EP2571510B1 - Biosynthetic proline/alanine random coil polypeptides and their uses - Google Patents

Biosynthetic proline/alanine random coil polypeptides and their uses Download PDF

Info

Publication number
EP2571510B1
EP2571510B1 EP11725656.0A EP11725656A EP2571510B1 EP 2571510 B1 EP2571510 B1 EP 2571510B1 EP 11725656 A EP11725656 A EP 11725656A EP 2571510 B1 EP2571510 B1 EP 2571510B1
Authority
EP
European Patent Office
Prior art keywords
amino acid
polypeptide
random coil
acid sequence
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP11725656.0A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP2571510A1 (en
Inventor
Arne Skerra
Uli Binder
Martin Schlapschy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Technische Universitaet Muenchen
XL Protein GmbH
Original Assignee
Technische Universitaet Muenchen
XL Protein GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Technische Universitaet Muenchen, XL Protein GmbH filed Critical Technische Universitaet Muenchen
Priority to EP11725656.0A priority Critical patent/EP2571510B1/en
Priority to SI201131597T priority patent/SI2571510T1/sl
Priority to EP18184601.5A priority patent/EP3473261A1/en
Priority to PL11725656T priority patent/PL2571510T3/pl
Publication of EP2571510A1 publication Critical patent/EP2571510A1/en
Application granted granted Critical
Publication of EP2571510B1 publication Critical patent/EP2571510B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/14Antitussive agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/001Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormone [GH], i.e. somatotropin
    • C07K14/615Extraction from natural sources
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor

Definitions

  • the present invention relates to a biosynthetic random coil polypeptide or a biosynthetic random coil polypeptide segment or a conjugate, said biosynthetic random coil polypeptide or a biosynthetic random coil polypeptide segment or a conjugate comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least about 50 proline (Pro) and alanine (Ala) amino acid residues.
  • Said at least about 50 proline (Pro) and alanine (Ala) amino acid residues may be (a) constituent(s) of a heterologous polypeptide or an heterologous polypeptide construct.
  • biosynthetic random coil polypeptides said polypeptide segments or said conjugates are described.
  • the uses may, inter alia, comprise medical uses, diagnostic uses or uses in the food industry as well as other industrial applications, like in the paper industry, in oil recovery and the like.
  • the present invention relates, also, to (a) specific use(s) of the herein provided biosynthetic random coil polypeptide or biosynthetic random coil polypeptide segment or conjugates, said biosynthetic random coil polypeptide or biosynthetic random coil polypeptide segment or conjugates comprising an amino acid sequence consisting solely of proline and alanine amino acid residues.
  • the amino acid sequence of the herein provided biosynthetic random coil polypeptide or biosynthetic random coil polypeptide segment consists of at least about 50, of at least about 100, of at least about 150, of at least about 200, of at least about 250, of at least about 300, of at least about 350 or of at least about 400 proline (Pro) and alanine (Ala) amino acid residues.
  • Said at least about 50, at least about 100, at least about 150, at least about 200, at least about 250, at least about 300, at least about 350 or at least about 400 proline (Pro) and alanine (Ala) amino acid residues are a constituent of a drug conjugate, like a conjugate with a biologically active compound.
  • heterologous proteins are provided herein whereby these proteins comprise at least two domains, wherein a first domain of said at least two domains comprises an amino acid sequence having and/or mediating an activity, like a biological activity, and a second domain of said at least two domains comprising the biosynthetic random coil proline/alanine polypeptide or proline/alanine polypeptide segment of the present invention.
  • the present invention relates in particular to a drug conjugate comprising (i) a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues, and (ii) a pharmaceutical drug selected from the group consisting of (a) a biologically active protein or a polypeptide that comprises or that is an amino acid sequence that has or mediates a biological activity and (b) a small molecule drug.
  • a further subject of the present invention is a drug conjugate comprising the biosynthetic random coil proline/alanine polypeptide or proline/alanine polypeptide segment as provided herein and, additionally, (a) pharmaceutically useful small molecule(s) or protein(s) linked and/or coupled to said biosynthetic random coil proline/alanine polypeptide or proline/alanine polypeptide segment.
  • nucleic acid molecules encoding the biosynthetic random coil polypeptide or polypeptide segment and/or the biologically active, heterologous proteins as well as vectors and cells comprising said nucleic acid molecules are disclosed.
  • compositions comprising the drug conjugates or the nucleic acid molecules, vectors and cells of the invention.
  • biosynthetic random coil polypeptides or polypeptide segments as well as of producing and/or obtaining the inventive biologically active, heterologous proteins, and/or polypeptide constructs or drug conjugates are provided.
  • medical, pharmaceutical as well as diagnostic uses are provided herein for the biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues (or for molecules and conjugates comprising the same) as defined herein.
  • Such a medical or pharmaceutical use can comprise the use of said biosynthetic random coil polypeptide or polypeptide segment as plasma expander and the like.
  • the means and methods provided herein are not limited to pharmaceutical, medical and biological uses but can also be employed in other industrial areas, like in the paper industry, in oil recovery, etc. Rapid clearance from blood circulation by renal filtration is a typical property of small molecules (including small proteins and peptides). However, by expanding the apparent molecular dimensions beyond the pore size of the kidney glomeruli plasma half-life of therapeutic proteins can be extended to a medically useful range of several days.
  • One strategy to achieve such an effect is chemical conjugation of the biologic with the synthetic polymer poly-ethylene glycol (PEG).
  • PEG-interferon alpha2a PEG-interferon alpha2a
  • PEG-G-CSF Neurogena®
  • Cimzia® PEGylated alphaTNF-Fab fragment
  • PEG is not biodegradable, which can cause side effects such as vacuolation of kidney epithelium upon continuous treatment; see, e.g., Gaberc-Porekar (2008) Curr Opin Drug Discov Devel 11:242-50 ; Knop (2010) Angew Chem Int Ed Engl 49:6288-308 or Armstrong in: Veronese (Ed.), "PEGylated Protein Drugs: Basic Science and Clinical Applications”; Birkhäuser Verlag, Basel 2009 .
  • PK studies in mice of the trans-sialidase containing 13 hydrophilic and (at physiological pH) negatively charged corresponding amino acid repeats having the natural sequence DSSAHSTPSTPA revealed a five-fold longer plasma half-life compared to the recombinant enzyme from which the C-terminal repetitive sequence had been deleted ( Buscaglia (1999) Blood 93 :2025-32 ).
  • a similar half-life extending effect was observed after fusion of the same trans-sialidase, i.e. its 76 kDa catalytic domain, with 13 charged amino acid repeats of the sequence EPKSA that were found in the Trypanosoma cruzi protein antigen 13.
  • Gelatin hydrolyzed and denatured animal collagen
  • Succinylation of gelatin primarily via the ⁇ -amino groups of naturally interspersed lysine side chains, increases the hydrophilicity of this biopolymer and lowers its isoelectric point (p I ).
  • the intramolecular electrostatic repulsion between the negatively charged carboxylate groups of the modified side chains supposedly spreads out the molecule into a more or less extended conformation.
  • the resulting expanded volume makes succinylated gelatin a macromolecule for use as plasma expander in humans and is, inter alia, marketed as Volplex® (Beacon Pharmaceuticals Ltd) or Gelofusine® (B. Braun Melsungen AG). Furthermore, a half-life extending effect was achieved by genetic fusion of granulocyte-colony-stimulating factor (G-CSF) to an artificial gelatin-like polypeptide ( Huang (2010) Eur J Pharm Biopharm 74:435-41 ).
  • G-CSF granulocyte-colony-stimulating factor
  • GLK gelatin-like protein
  • G-CSF was fused at its N-terminus with 4 copies of this GLK sequence and secreted in Pichia pastoris.
  • Pichia pastoris appeared as a favourable production organism for GLK fusion proteins; yet, if GLKs can also be produced in other organisms remains to be determined as it is known that recombinant gelatin fragments can be expressed with only low yield in E. coli, for example, as illustrated in Olsen (2003), Adv Drug Deliv Rev 55:1547-67 .
  • Elastin is a component of the extracellular matrix in many tissues. It is formed from the soluble precursor tropoelastin, which consists of a hydrophilic Lys/Ala-rich domain and a hydrophobic, elastomeric domain with repetitive sequence. Enzymatic crosslinking of lysine side chains within the hydrophilic domain leads to insoluble elastin formation. Elastin-like polypeptides (ELPs) are artifically designed, repetitive amino acid sequences derived from the hydrophobic domain of tropoelastin.
  • ELPs Elastin-like polypeptides
  • ELPs The most common repeat sequence motif of ELPs is V-P-G-X-G, wherein "X" can be any amino acid except Pro ( MacEwan (2010) Biopolymers 94:60-77 ; Kim (2010) Adv Drug Deliv Rev 62:1468-78 ).
  • Suitable ELPs can be fused with therapeutic proteins and produced in E. coli. Consequently, the ability of ELPs to form gel-like depots after injection can significantly prolong the in vivo half-life of an attached biologic, albeit by a mechanism different from the other unstructured polypeptides.
  • ELP attachment can hamper the bioactivity of the fusion partner as demonstrated for the interleukin-1 receptor antagonist in an IL-1-induced lymphocyte proliferation bioassay ( Shamji (2007) Arthritis Rheum. 11:3650- 3661 ).
  • ELPs are subject to degradation by endogenous proteases such as collagenase.
  • aggregated proteins are generally more susceptible to immunogenicity.
  • polyglutamate has been chemically coupled to poorly soluble cytotoxic small molecule drugs for cancer treatment.
  • a corresponding product would be OpaxioTM, a paclitaxel drug conjugate currently in clinical phase III studies.
  • Half-life of a paclitaxel PG conjugate was prolonged by a factor 3 to 14 in comparison with the unmodified compound ( Singer (2005) J Control Release 109:120-6 ).
  • Further fusion proteins for example G-CSF fused at its N-terminus with a stretch of 175 consecutive Glu residues or IFN-alpha2 carrying at its C-terminus a PG tail of 84 residues, were produced in a soluble state in the cytoplasm of E.
  • WO 2006/081249 describes a polypeptide sequence with about 2 to 500 repeat units of 3 to 6 amino acids, wherein G, N or Q represent the major constituents while minor constituents can be A, S, T, D or E.
  • This amino acid composition allows integration of the glycosylation sequon Asn-Xaa-Ser/Thr (where Xaa is any amino acid except Pro) for N-linked glycosylation of the Asn side chain in eukaryotic expression systems.
  • the increased macromolecular size of a resulting fusion protein, including posttranslational modification with bulky solvated carbohydrate structures, can extend the pharmacokinetics of the genetically conjugated protein.
  • oligosaccharide attachments in general can both reduce susceptibility to proteolysis and increase the hydrodynamic volume ( Sinclair (2005) J Pharm Sci 94:1626-35 ).
  • a disadvantage is the intrinsic molecular heterogeneity of the glycosylated biomacromolecule, which causes additional effort during biotechnological production and quality control.
  • WO 2010/091122 and WO 2007/103515 ) and Schellenberger (2009) Nat Biotechnol 27:1186-90 disclose unstructured non-repetitive amino acid polymers encompassing and comprising the residues P, E, S, T, A and G.
  • This set of amino acids which shows a composition not unlike the PSTAD repeat described further above, was systematically screened for sequences to yield a solvated polypeptide with large molecular size, suitable for biopharmaceutical development, by avoiding hydrophobic side chains - in particular F, I, L, M, V and W - that can give rise to aggregation and may cause an HLA/MHC-II mediated immune response.
  • glucagon XTEN fusion showed merely 15 % bioactivity of the non-modified peptide.
  • An even stronger loss in receptor affinity (17-fold increased EC 50 ) was described for an XTEN fusion of human growth hormone (hGH); see WO 2010/144502
  • hGH human growth hormone
  • glycine as the smallest and structurally simplest amino acid, has been considered as the conformationally most flexible amino acid based on theoretical grounds; see, e.g. Schulz GE, Schirmer RH. Principles of Protein Structure. Springer, New York 1979 .
  • polyglycine is a linear unbranched polyamide that shows certain resemblance to the polyether PEG in so far as both are essentially one-dimensional macromolecules with many rotational degrees of freedom along the chain, which are made of repeated short hydrocarbon units that are regularly interrupted by hydrogen-bonding and highly solvated polar groups. Consequently, polyglycine should constitute the simplest genetically encodable PEG mimetic with prospects for extending the plasma half-life of therapeutic proteins.
  • HAP homo-amino-acid polymer
  • GRS glycine rich sequence
  • WO 2008/155134 discloses that sequences with an appropriate mixture of Pro, Ala, and Ser (i.e. PAS) residues lead to mutual cancellation of their distinct secondary structure preferences and, thus, result in a stably disordered polypeptide.
  • PAS Pro, Ala, and Ser residues
  • fusion proteins with a domain composed only of serine and alanine (SA) residues, i.e. a domain comprising only two types of amino acids do not form a random coil, but a ⁇ -sheet structure instead.
  • SA serine and alanine
  • US 2009/0192087 discloses a poly-Ala sequence which serves as a linker to fuse at least one extracellular subdomain of the Klotho protein with a polypeptide comprising a fibroblast growth hormone.
  • EP-A1 1 739 167 discloses a method for the generation of viruses and their use in the preparation of vaccines.
  • EP-A1 1 739 167 also discloses a poly-A-sequence of a polypeptide that can be used for preparing viruses. Yet, these poly-A-sequences do not consist solely of proline and alanine residues.
  • WO 2008/14500 discloses parvovirus-proteins with certain insertions and their medical use. Yet, WO 2008/14500 neither discloses a protein having an amino acid sequence consisting solely of at least 50 proline and alanine residues nor a drug conjugate comprising same.
  • WO 01/78503 discloses GFP-fusion proteins for expression in plant cells, wherein the fusion proteins can comprise a (hydroxy)proline-alanine sequence. Yet, GFP is not a pharmaceutical drug.
  • copolypeptides of proline and alanine are partially soluble in water, while other copolypeptides were completely insoluble. It is speculated in Izuka that proline/alanine copolypeptides may have a partial disordered conformation. Izuka emphasizes that chemically synthesized polypeptides with a random proline/alanine sequence occur predominantly in a collagen-like conformation, i.e. in a structured conformation.
  • the technical problem underlying the present invention is the provision of large polypeptides with true random coil conformation.
  • the technical problem is solved by provision of the embodiments characterized in the claims and as provided herein.
  • the invention relates to a drug conjugate comprising (i) a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues, and (ii) a pharmaceutical drug selected from the group consisting of (a) a biologically active protein or a polypeptide that comprises or that is an amino acid sequence that has or mediates a biological activity and (b) a small molecule drug.
  • polypeptides with true random coil conformation and polypeptide segments with true random coil conformation as provided herein are also useful in the context of cosmetic uses as well as uses in food industry and the production of beverages.
  • the large polypeptides provided herein which show true random coil confirmation consist soley and merely of proline (P, Pro) and alanine (A, Ala) residues and comprise more than at least 50 amino acids, in particular of at least about 100, in particular of at least about 150, in particular of at least about 200, in particular of at least about 250, in particular of at least about 300, in particular of at least about 350, in particular of at least about 400 proline and alanine amino acid residues.
  • nucleic acid molecules that encode for the herein disclosed biosynthetic random coil polypeptides or polypeptide segments as well as for drug or food conjugates that comprise said biosynthetic random coil polypeptides or polypeptide segments and a (covalently linked) protein of interest, like a biologically active protein.
  • biosynthetic random coil polypeptide or biosynthetic random coil polypeptide segment as described herein and to be used in drug or food conjugates as provided herein and comprising an amino acid sequence consisting of at least about 50, of at least about 100, of at least about 150, of at least about 200, of at least about 250, of at least about 300, of at least about 350, of at least about 400 proline (P) and alanine (A) amino acid residues is, inter alia, to be used in a heterologous context, i.e.
  • a biologically active heterologous protein, protein construct and/or in a drug conjugate comprising said biosynthetic random coil polypeptide or polypeptide segment and pharmaceutically or medically useful molecules, like small molecules, peptides or biomacromolecules such as proteins, nucleic acids, carbohydrates, lipid vesicles and the like.
  • pharmaceutically or medically useful molecules like small molecules, peptides or biomacromolecules such as proteins, nucleic acids, carbohydrates, lipid vesicles and the like.
  • the inventors could successfully provide for drug conjugates which consist of the true random coil polypeptides as defined herein and biologically active proteins or protein stretches as well as drug conjugates that consist of small molecules or small molecule drugs that comprise and/or are linked to the herein described random coil polypeptides, consisting solely of proline and alanine amino acid residues (i.e. of both amino acids P and A)
  • the present invention provides, inter alia, for a biologically active, heterologous protein comprising at least two domains wherein (a) a first domain of said at least two domains comprises an amino acid sequence having and/or mediating said biological activity; and (b) a second domain of said at least two domains comprises the biosynthetic random coil polypeptide or polypeptide segment consisting of an amino acid sequence consisting of at least about 50, of at least about 100, of at least about 150, of at least about 200, of at least about 250, of at least about 300, of at least about 350, of at least about 400 proline and alanine amino acid residues.
  • said "first domain and said "second domain” are not comprised in either a natural (i.e. occurring in nature) protein or a hypothetical protein as deduced from naturally occurring coding nucleic acid sequences, like open reading frames etc.
  • this invention provides for a drug conjugate consisting of the biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting of at least about 50, of at least about 100, of at least about 150, of at least about 200, of at least about 250, of at least about 300, of at least about 350, of at least about 400 proline and alanine amino acid residues and (a) pharmaceutically useful molecule(s), like (a) small molecule(s) or (a) protein(s) that is/are conjugated to said biosynthetic random coil polypeptide or polypeptide segment.
  • biologically active in context of herein disclosed conjugates is not limited to pure biological molecules but also comprise medically active, therapeutically active, pharmaceutically active molecules and the like. It is evident for the skilled artisan that the means and methods provided herein are not limited to pharmaceutical and medical uses, but can be employed in a wide variety of technologies, including, but not limited to cosmetic, food, beverage and nutrition technologies, oil industry, paper industry and the like.
  • the random coil polypeptides provided herein are biosynthetically produced.
  • biosynthetic refers to the synthesis by means of biotechnological methods (in contrast to chemical synthesis). Such biotechnological methods are well known in the art and also described herein further below.
  • the biosynthesis of the random coil polypeptides of the present invention allows the production of polypeptides with a defined sequence of proline and alanine residues, a defined length and/or a defined ratio of proline and alanine residues.
  • the polypeptides provided in accordance with the present invention are substantially pure, i.e.
  • the produced polypeptides are essentially uniform and share the above characteristics (i.e. defined sequence, defined length and/or defined amino acid ratio).
  • the random coil polypeptides consisting of at least about 50, in particular of at least about 100, in particular of at least about 150, in particular of at least about 200, in particular of at least about 250, in particular of at least about 300, in particular of at least about 350, in particular of at least about 400 proline and alanine amino acid residues are in accordance with this invention comprised in drug conjugates.
  • the above features of the polypeptides of the present invention permit the formation of a stable random coil of the polypeptides and these random coil polypeptides have surprising and advantageous properties.
  • the polypeptides of the present invention are completely soluble in aqueous solution and have an increased hydrodynamic volume.
  • the random coil polypeptides as defined herein are also capable of conferring an increased in vivo / in vitro stability. This is particularly important for medical applications, for example, for biologically active proteins or drug conjugates comprising the random coil polypeptide of this invention.
  • the numerous advantageous properties of the random coil polypeptides of the present invention not only permit their use in the medical field but also in other fields, like in cosmetics/cosmetic treatments or in the fields of nutrition and food technology, like in the dairy industry or in meat processing.
  • Examples of conjugates useful in food industry and the like are conjugates that comprise the herein disclosed random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues and compounds that are useful in these technologies, like, e.g. polyoxypropylene or polyoxyethylene polymers, which are non-ionic surfactants used as emulsifiers.
  • biosynthetic random coil polypeptide as defined herein in biochemical methods and in technical processes, such as paper production, oil recovery and the like.
  • biosynthetic random coil polypeptides consisting merely of proline and alanine residues as provided herein (and as also of the herein disclosed conjugates and constructs, like drug or food conjugates/constructs, comprising said biosynthetic, true random coil polypeptides) are described below in greater detail.
  • illustrative uses and means and methods employing these inventive biosynthetic random coil polypeptides are provided below.
  • proline-alanine polymers/polypeptides with a uniform composition form stable random coil conformation. This is also demonstrated in the appended examples, where random coil structure of biosynthetic proline/alanine (co)-polymers/polypeptides is confirmed by circular dichroism (CD) spectroscopy.
  • CD circular dichroism
  • the chemically synthesized polypeptides described, for example, in Izuka (1993), loc. cit. have an arbitrary/undefined and stochastic sequence and a diverse length.
  • the chemically synthesized polypeptides comprise a mixture of completely different peptides with various proline/alanine ratios, lengths, and so on.
  • the chemically synthesized polypeptides of such a mixture do not (or only partially) form a random coil and, accordingly, do not have any of the advantageous properties of the biosynthetic polypeptides provided and described herein below.
  • the present disclosure comprises and relates to compositions comprising the inventive biosynthetic random coil polypeptides/polymers as disclosed herein, whereby said biosynthetic random coil polypeptides/polymers are defined, inter alia, by their sequence comprising solely proline and alanine residues.
  • the invention is defined by the appended claims.
  • the invention relates to drug conjugates comprising, as one constituent, these random coil polypeptides/polymers disclosed herein.
  • These inventive biosynthetic random coil polypeptides/polymers comprised in said compositions are, in one embodiment, of uniform length.
  • the biosynthetic random coil polypeptides (or random coil polypeptide segments) of this invention consisting solely of proline and alanine residues unexpectedly form a stable random coil conformation.
  • random coil as used herein relates generally to any conformation of a polymeric molecule, including amino acid polymers/amino acid sequences/polypeptides, in which the individual monomeric elements that form said polymeric structure are essentially randomly oriented towards the adjacent monomeric elements while still being chemically bound to said adjacent monomeric elements.
  • a polypeptide, amino acid sequence or amino acid polymer adopting/having/forming "random coil conformation" substantially lacks a defined secondary and tertiary structure.
  • the monomeric elements forming the polymeric structure are either single amino acids such as proline and alanine per se or peptide stretches such as the "amino acid repeats"/"amino acid cassettes"/"cassette repeats"/"building blocks"/"modules" (or fragments thereof) which are described and defined further below.
  • polypeptide random coils and their methods of experimental identification are known to the person skilled in the art and have been described in the scientific literature ( Cantor (1980) Biophysical Chemistry, 2nd ed., W. H. Freeman and Company, New York ; Creighton (1993) Proteins - Structures and Molecular Properties, 2nd ed., W. H. Freeman and Company, New York ; Smith (1996) Fold Des 1:R95-R106 ).
  • segment refers to a part of the herein defined biosynthetic random coil polypeptide, whereby such a part may be an internal part of the biosynthetic random coil polypeptide described herein.
  • Such a “segment” may be, for example, a biosynthetic random coil polypeptide as defined herein where one (or more) amino acid(s) has/have been deleted, e.g. from the start and/or from the end of the polypeptide of the invention.
  • a “segment” may be used as or may form part of a larger protein or polypeptide, for example, of a fusion protein with a biologically active protein.
  • fusion protein would also be an example of a heterologous, biologically active polypeptide/protein/polypeptide construct of the present invention.
  • heterologous as used herein is defined herein below.
  • the random coil polypeptide (or random coil segment thereof), as provided in the present invention and to be employed in context of this invention, adopts/forms random coil conformation, for example, in aqueous solution or at physiological conditions.
  • physiological conditions is known in the art and relates to those conditions in which proteins usually adopt their native, folded conformation. More specifically, the term “physiological conditions” relates to the biophysical parameters as they are typically valid for higher forms of life and, particularly, in mammals, most preferably human beings.
  • the term “physiological conditions” may relate to the biochemical and biophysical parameters as they are normally found in the body (in particular in body fluids) of mammals and in particular in humans.
  • Said “physiological conditions” may relate to the corresponding parameters found in the healthy body as well as the parameters found under disease conditions or in human patients. For example, a sick mammal or human patient may have a higher, yet “physiological” temperature condition when said mammal or said human suffers from fever. With respect to “physiological conditions” at which proteins adopt their native conformation/state, the most important parameters are temperature (37 °C for the human body), pH (7.35 - 7.45 for human blood), osmolarity (280 - 300 mmol/kg H 2 O), and, if necessary, protein content (66 - 85 g/l serum). Yet, the person skilled in the art is aware that at physiological conditions these parameters may vary, e.g.
  • the temperature, pH, osmolarity, and protein content may be different in given body or tissue fluids such as blood, liquor cerebrospinalis, peritoneal fluid and lymph ( Klinke (2005) Physiologie, 5th ed., Georg Thieme Verlag, Stuttgart ).
  • body or tissue fluids such as blood, liquor cerebrospinalis, peritoneal fluid and lymph ( Klinke (2005) Physiologie, 5th ed., Georg Thieme Verlag, Stuttgart ).
  • the osmolarity may be around 290 mmol/kg H 2 O and the protein concentration may be between 0.15 g/l to 0.45 g/l while in the lymph the pH may be around 7.4 and the protein content may be between 3 g/l and 5 g/l.
  • the biophysical parameters such as temperature, pH, osmolarity and protein content may be different to the physiological conditions normally found in vivo. Temperatures between 1 °C and 42 °C or preferably 4 °C to 25 °C may be considered useful to test and/or verify the biophysical properties and biological activity of a protein under physiological conditions in vitro.
  • buffers in particular in experimental settings (for example in the determination of protein structures, in particular in CD measurements and other methods that allow the person skilled in the art to determine the structural properties of a protein/amino acid stretch) or in buffers, solvents and/or excipients for pharmaceutical compositions, are considered to represent "physiological solutions" / "physiological conditions" in vitro.
  • buffers are, e.g. phosphate-buffered saline (PBS: 115 mM NaCl, 4 mM KH 2 PO 4 , 16 mM Na 2 HPO 4 pH 7.4), Tris buffers, acetate buffers, citrate buffers or similar buffers such as those used in the appended examples.
  • the pH of a buffer representing "physiological solution conditions” should lie in a range from 6.5 to 8.5, preferably in a range from 7.0 to 8.0, most preferably in a range from 7.2 to 7.7 and the osmolarity should lie in a range from 10 to 1000 mmol/kg H 2 O, more preferably in a range from 50 to 500 mmol/kg H 2 O and most preferably in a range from 200 to 350 mmol/kg H 2 O.
  • the protein content of a buffer representing physiological solution conditions may lie in a range from 0 to 100 g/l, neglecting the protein with biological activity itself, whereby typical stabilizing proteins may be used, for example human or bovine serum albumin.
  • aqueous solution may be a solution with a water (H 2 O) content of at least about 20 %, of at least about 30 %, of at least about 40 %, of at least about 50 %, of at least about 60 %, of at least about 70 %, of at least about 80 % or of at least about 90 % H 2 O (weight/weight).
  • H 2 O water
  • the polypeptide (or segment thereof) of the present invention may form random coil conformation in aqueous solution, possibly containing other miscible solvents, or in aqueous dispersions with a wider range of temperatures, pH values, osmolarities or protein content. This is particularly relevant for applications of the random coil polypeptide (or segment thereof) outside medical therapy or in vivo diagnostics, for example in cosmetics, nutrition or food technology.
  • the random coil conformation of the proline/alanine biosynthetic polypeptide (or segment thereof) of the present invention is maintained in and/or is used in context of pharmaceutical compositions, like liquid pharmaceuticals/biologicals or lyophilized pharmaceutical compositions.
  • pharmaceutical compositions like liquid pharmaceuticals/biologicals or lyophilized pharmaceutical compositions.
  • drug conjugates comprising, inter alia, the inventive random coil polypeptide (or polypeptide segment).
  • physiological conditions are to be used in corresponding buffer systems, solvents and/or excipients.
  • lyophilized or dried compositions like, e.g.
  • random coil conformation of the herein provided random coil polypeptide (or polypeptide segment) is transiently not present and/or cannot be detected.
  • said random coil polypeptide (or polypeptide segment) will adopt/form again its random coil after reconstitution in corresponding buffers/solutions/excipients/solvents or after administration to the body.
  • Methods for determining whether a polypeptide (or segment thereof) forms/adopts random coil conformation are known in the art (Cantor (1980) loc. cit.; Creighton (1993) loc. cit.; Smith (1996) loc. cit.).
  • Such methods include circular dichroism (CD) spectroscopy as exemplified herein below.
  • CD spectroscopy represents a light absorption spectroscopy method in which the difference in absorbance of right- and left-circularly polarized light by a substance is measured.
  • the secondary structure of a protein can be determined by CD spectroscopy using far-ultraviolet spectra with a wavelength between approximately 190 and 250 nm. At these wavelengths, the different secondary structures commonly found in polypeptides can be analyzed, since ⁇ -helix, parallel and anti-parallel ⁇ -sheet, and random coil conformations each give rise to a characteristic shape and magnitude of the CD spectrum.
  • CD spectrometry the skilled artisan is readily capable of determining whether polypeptide (or segment thereof) forms/adopts random coil conformation in aqueous solution or at physiological conditions.
  • Other established biophysical methods include nuclear magnetic resonance (NMR) spectroscopy, absorption spectrometry, infrared and Raman spectroscopy, measurement of the hydrodynamic volume via size exclusion chromatography, analytical ultracentrifugation or dynamic/static light scattering as well as measurements of the frictional coefficient or intrinsic viscosity (Cantor (1980) loc. cit.; Creighton (1993) loc. cit.; Smith (1996) loc. cit.).
  • Homo-polymers of most amino acids are usually insoluble in aqueous solution ( Bamford (1956) Synthetic Polypeptides - Preparation, Structure, and Properties, 2nd ed., Academic Press, New York ).
  • Homo-polymers of several hydrophilic amino acids are known to form secondary structures, for example ⁇ -helix in the case of Ala ( Shental-Bechor (2005) Biophys J 88:2391-2402 ) and ⁇ -sheet in the case of Ser ( Quadrifoglio (1968) J Am Chem Soc 90:2760-2765 ) while poly-proline, the stiffest homooligopeptide ( Schimmel (1967) Proc Natl Acad Sci USA 58:52-59 ), forms a type II trans helix in aqueous solution ( Cowan (1955) Nature 176:501-503 ).
  • amino acid sequences comprising proline residues are expected to have a relatively compact hydrodynamic volume.
  • hydrodynamic volume of the amino acid polymers/polypeptides of the invention that comprise a mixture of proline and alanine residues have a dramatically increased hydrodynamic volume as determined by analytical gel permeation / size exclusion chromatography when compared to the expected hydrodynamic volume.
  • polypeptides comprising mixtures of these two amino acids (proline and alanine) of which each alone tends to form a homooligopeptide with defined secondary structure, adopt random coil conformation under physiological conditions.
  • Such inventive proline/alanine polypeptides have a larger hydrodynamic radius than homo-polymers comprising the same number of Gly residues, for example, and they confer better solubility to the biologically active proteins or constructs, i.e. biologically active heterologous proteins or drug conjugates, according to the invention.
  • biosynthetic random coil proline/alanine polypeptides of the present invention differ from chemically synthesized polypeptides in that they can adopt a defined, uniform length by easy means and methods.
  • the prior art provides mixtures/compositions of polypeptides with enormous variations in terms of the length of the peptides
  • the present invention can provide mixtures/compositions of biosynthetic random coil polypeptides with a defined length.
  • essentially all polypeptides of the invention comprised in such a mixture/composition have the same defined length, and, hence, share the same biochemical characteristics.
  • biosynthetic random coil polypeptides can be employed.
  • biosynthetic random coil polypeptides or polypeptide segments is, accordingly, the use as plasma expander.
  • drug conjugates like heterologous polypeptides or heterologous polypeptide constructs may be employed in context of the medical or pharmaceutical intervention of a disorder related to an impaired blood plasma amount or blood plasma content or of a disorder related to an impaired blood volume.
  • the present invention relates in one embodiment to a biosynthetic random polypeptide (or segment thereof) which comprises an amino acid sequence consisting solely of at least about 50 proline and alanine amino acid residues, of at least about 100 proline and alanine amino acid residues, of at least about 150 proline and alanine amino acid residues or of at least about 200 proline and alanine residues when comprised in a drug conjugate, like a heterologous protein/polypeptide/polypeptide construct.
  • the present invention also relates to biosynthetic random coil polypeptides which comprise an amino acid sequence consisting solely of at least about 200 proline and alanine amino acid residues, even more preferably of at least about 300 proline and alanine amino acid residues, particularly preferably of at least about 400 proline and alanine amino acid residues, more particularly preferably of at least about 500 proline and alanine amino acid residues and most preferably of at least about 600 proline and alanine amino acid residues to be used herein.
  • the amino acid sequence forming random coil conformation may consist of maximally about 3000 proline and alanine amino acid residues, of maximally about 2000 proline and alanine amino acid residues, of maximally about 1500 proline and alanine amino acid residues, of maximally about 1200 proline and alanine amino acid residues, of maximally about 800 proline and alanine amino acid residues.
  • the proline/alanine amino acid sequence stretch may consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400, of about 500, of about 600, of about 700, of about 800, of about 900 to about 3000 proline and alanine amino acid residues.
  • the inventive biosynthetic amino acid sequence to be used herein comprises about 200 to about 3000 proline and alanine residues, about 200 to about 2500 proline and alanine residues, about 200 to about 2000 proline and alanine residues, about 200 to about 1500 proline and alanine residues, about 200 to about 1000 proline and alanine residues, about 300 to about 3000 proline and alanine residues, about 300 to about 2500 proline and alanine residues, about 300 to about 2000 proline and alanine residues, about 300 to about 1500 proline and alanine residues, about 300 to about 1000 proline and alanine residues, about 400 to about 3000 proline and alanine residues, about 400 to about 2500 proline and alanine residues, about 400 to about 2000 proline and alanine residues, about 400 to about 1500 proline and alanine residues, about 400 to about 1000 proline and alanine residues, about 500 to about 3000 pro
  • biosynthetic amino acid sequences consisting essentially of proline and alanine
  • biosynthetic amino acid sequences are within the scope of this invention and can readily be employed in the herein defined biologically active proteins or protein constructs which comprise as one domain of at least two domains an amino acid sequence having and/or mediating said biological activity and as another domain of at least two domains the biosynthetic random coil polypeptide or polypeptide segment consisting of at least about 50 proline and alanine amino acid residues, of at least about 100 proline and alanine amino acid residues, of at least about 150 proline and alanine amino acid residues, of at least about 200, of at least about 250, of at least about 300, of at least about 350, of at least about 400 proline and alanine amino acid residues.
  • biosynthetic random coil polypeptide or polypeptide segment corresponds to the biosynthetic random coil part of a heterologous protein/protein construct.
  • biosynthetic proline/alanine stretches consist of maximally about 3000 proline and alanine amino acid residues.
  • These amino acid sequences comprise proline and alanine as main or unique residues as explained further below.
  • biosynthetic amino acid sequence consisting solely of proline (P) and alanine (A) amino acid residues, which forms/adopts/has a random coil conformation.
  • biosynthetic polypeptide or polypeptide seqment consists of the amino acid sequence having a random coil conformation as defined herein.
  • biosynthetic polypeptide may, in addition to the herein described amino acid sequence forming/adopting/having a random coil conformation, comprise further amino acid sequences/amino acid residues which do not contribute to the formation of the random coil conformation or which are not capable of forming/adopting/having a random coil conformation on their own.
  • biosynthetic polypeptides or segments thereof are biosynthetic "random coil" polypeptides or polypeptide segments.
  • the further amino acid sequences/amino acid residues may, for example, be useful as linkers. Inter alia, dimers, trimers, i.e.
  • multimers of the biosynthetic random coil polypeptide are also envisaged in context of the present invention and such multimers may be linked by amino acid sequences/residues which do not form random coil conformation.
  • An example of a protein which may comprise such a random coil polypeptide is the herein provided biologically active protein, which may, in addition to the random coil polypeptide consisting of proline and alanine amino acid residues as defined herein further comprise another polypeptide having/mediating biological activity.
  • such a construct may be a heterologous, biologically active protein or polypeptide construct as described herein.
  • amino acid residues is not limited to the concise number of amino acid residues but also comprises amino acid stretches that comprise either additional about 1-20 %, like 10 % to 20 % residues or about 1-20 %, like about 10 % to 20 % less residues.
  • amino acid residues may also comprise about 80 to 100 and about 100 to 120 amino acid residues without deferring from the gist of the present invention.
  • at least about 200 amino acid residues may also comprise about 160 to 200 and about 200 to 240 amino acid residues without deferring from the gist of this invention.
  • biosynthetic random coil polypeptides are characterised by a defined content or ratio of amino acid residues, in particular of the main constituents proline and alanine.
  • the present invention relates to a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least about 50, of at least about 100, of at least about 150, of at least about 200 of at least about 250, of at least about 300, of at least about 350, of at least about 400 proline (Pro) and alanine (Ala) amino acid residues in particular when comprised in a heterologous biological active protein/protein construct/polypeptide or drug conjugate.
  • the present invention relates in one embodiment to a biosynthetic random coil polypeptide (or segment thereof) whereby the amino acid sequence consists mainly of proline and alanine, and wherein the proline residues constitute more than about 10 % and less than 75 % of the amino acid sequence.
  • the alanine residues comprise the remaining at least 25 % to 90 % of said amino acid sequence (or the random coil polypeptide or polypeptide segment if it consists of the amino acid sequence).
  • the amino acid sequence comprises more than about 10 %, preferably more than about 12 %, even more preferably more than about 14 %, particularly preferably more than about 18 %, more particularly preferably more than about 20 %, even more particularly preferably more than about 22 %, 23 % or 24 % and most preferably more than about 25 % proline residues.
  • the amino acid sequence preferably comprises less than about 75 %, more preferably less than 70 %, 65 %, 60 %, 55 % or 50 % proline residues, wherein the lower values are preferred. Even more preferably, the amino acid sequence comprises less than about 48 %, 46 %, 44 %, 42 % proline residues.
  • amino acid sequences comprising less than about 41 %, 40 %, 39 % 38 %, 37 % or 36 % proline residues, whereby lower values are preferred.
  • amino acid sequence comprise less than about 35 % proline residues; see also the herein below provided PA constructs.
  • the amino acid sequence preferably comprises less than about 90 %, more preferably less than 88 %, 86 %, 84 %, 82 % or 80 % alanine residues, wherein the lower values are preferred. Even more preferably, the amino acid sequence comprises less than about 79 %, 78 %, 77 %, 76 % alanine residues, whereby lower values are preferred. Most preferably, the amino acid sequence comprises less than about 75 % alanine residues.
  • an amino acid sequence comprising more than about 25 %, preferably more than about 30 %, even more preferably more than about 35 %, particularly preferably more than about 40 %, more particularly preferably more than about 45 % or 50 %, even more particularly preferably more than about 52 %, 54 %, 56 %, 58 % or 59 % alanine residues, wherein the higher values are preferred.
  • the amino acid sequence comprises more than about 60 %, 61 %, 62 %, 63 % or 64 % alanine residues and most preferably more than about 65 % alanine residues.
  • the random coil polypeptide may comprise an amino acid sequence consisting of about 25 % proline residues, and about 75 % alanine residues.
  • the random coil polypeptide (or segment thereof) may comprise an amino acid sequence consisting of about 35 % proline residues and about 65 % alanine residues.
  • the term "about X %" as used herein above is not limited to the concise number of the percentage, but also comprises values of additional 10 % to 20 % or 10 % to 20 % less residues.
  • 10 % may also relate to 11 % or 12 % and to 9 % and 8 %, respectively.
  • a biosynthetic random coil polypeptide (or segment thereof)/the amino acid sequence consists exclusively of proline and alanine amino acid residues, i.e. no other amino acid residues are present in the random coil polypeptide or in the amino acid sequence.
  • the amino acid sequences/polypeptides adopting random coil conformation may comprise a plurality of "amino acid repeats"/"amino acid cassettes"/"cassette repeats", wherein said "amino acid repeats"/"amino acid cassettes"/"cassette repeats"/"building block”/”modules” (these terms are used herein interchangeably) exclusively consist of proline (Pro, P) and alanine (Ala, A) amino acid residues (depicted herein as "PA”, or as "AP”), wherein no more than 6 consecutive amino acid residues are identical.
  • proline Pro
  • Al alanine amino acid residues
  • An illustrative "building block” is, e.g. "AP” and this has also been provided in the appended illustrative examples as functional biosynthetic random coil domain of the present invention.
  • This illustrative example is the sequence "P1A1" as also provided in form of APAPAPAPAPAPAPAP (SEQ ID NO: 51). i.e. a "poly PA” "amino acid repeat”/"amino acid cassette”/"cassette repeat".
  • the amino acid sequence/polypeptide comprising the above defined “amino acid repeats"/"amino acid cassettes"/"cassette repeats" and the like comprises no more than 5 identical consecutive amino acid residues.
  • Other alternative embodiments are provided herein below in context of exemplified, individual building blocks.
  • amino acid repeats may be identical or non-identical.
  • amino acid repeats building blocks
  • modules modules
  • repeats amino acid cassettes
  • amino acid cassettes etc. consisting of proline and alanine residues
  • SEQ ID NO: 1 SEQ ID NO: 2
  • SEQ ID NO: 3 SEQ ID NO: 4
  • SEQ ID NO: 5 SEQ ID NO: 6
  • SEQ ID NO. 51 The enclosed sequence listing also comprises illustrative nucleic acid sequences which encode such "repeats"/"modules", etc.
  • “Fragments” of these sequences to be employed in accordance with this invention for the generation of the random coil polypeptide (or segment thereof) may consist of at least 3, preferably of at least 4, more preferably of at least 5, even more preferably of at least 6, still more preferably of at least 8, particularly preferably of at least 10, more particularly preferably of at least 12, even more particularly preferably of at least 14, still more particularly preferably of at least 16, and most preferably of at least 18 consecutive amino acids of the amino acid sequence selected from the group consisting of said SEQ ID NOs: 1, 2, 3, 4, 5, 6 and 51 (here it is of note that SEQ ID No. 51 consists of an illustrative "AP" or "PA” repeat).
  • random coil conformation forming amino acid sequences/polypeptides to be used as building blocks or modules of the herein defined random coil polypeptide (or segment thereof) may, inter alia, comprise combinations and/or fragments or circularly permuted versions of the specific "building blocks", "polymer cassettes", or "polymer repeats" shown above.
  • the exemplified modules/sequence units/polymer repeats/ polymer cassettes of the random coil polypeptide/amino acid sequence may also provide for individual fragments which may be newly combined to form further modules/sequence units/polymer repeats/polymer cassettes in accordance with this invention.
  • module(s) means for treating or preventing human diseases and conditions in accordance with the following abbreviations: "sequence unit(s)”, “polymer repeat(s)”, “polymer cassette(s)” and “building block(s) are used as synonyms herein and relate to individual amino acid stretches which may be used to form the herein defined random coil polypeptide (or segment thereof)/amino acid sequence.
  • amino acid repeat (used as "building block” etc. of a biosynthetic random coil polypeptide of the present invention) may consist of at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more amino acid residues, wherein each repeat comprises (an) proline and alanine residue(s).
  • said "building block” can also merely consist of the 2 herein provided amino acid residues P and A, namely in form of "PA” or "AP".
  • the amino acid repeat according to the present invention does not comprise more than 50 amino acid residues.
  • such a “repeat” may comprise even more than 50 amino acid residues, for example in cases wherein said inventive biosynthetic random coil polypeptide/polymer comprises more than about, e.g., 100 amino acids, more than about 150 amino acids, more than about 200 amino acids, etc. Accordingly, the maximal amount of amino acid residues comprised in such a "repeat” is conditioned by the over-all length of the biosynthetic polypeptide (or segment thereof)/polymer as provided herein.
  • biosynthetic random coil polypeptides/amino acid sequences comprising the above repeats etc. should preferably have the overall length and/or proline/alanine content as defined and explained herein above, i.e. consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 amino acids and/or comprise more than about 10 % and less than about 75 % proline residues. All the definitions given herein above in this context also apply here, mutatis mutandis.
  • the present invention provides for (a) biologically active, heterologous protein(s) or (a) protein construct(s) that is/are particularly useful in a pharmaceutical, medical and/or medicinal setting.
  • biologically active, heterologous proteins/protein constructs comprise as at least one domain of said at least two domains the random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine residues, wherein said amino acid sequence consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 proline (Pro) and alanine (Ala) residues.
  • heterologous relates to at least two domains within said proteins, polypeptides or protein constructs wherein a first of said at least two domains confers, has and/or mediates a defined biological activity and wherein a second of said at least two domains comprises the biosynthetic random coil polypeptide consisting solely of proline and alanine amino acid residues and whereby said at least two domains are not found operationally linked to each other in nature or are not encoded by a single coding nucleic acid sequence (like an open reading frame) existing in nature.
  • biosynthetic random coil polypeptide/polypeptide segment consisting solely of proline and alanine amino acid residues as provided herein and as employed in the biologically active, heterologous proteins/protein constructs of this invention are preferably not further (chemically) modified, for example they are preferably neither glycosylated nor hydroxylated.
  • such a homologous hypothetical protein as deduced from a naturally occurring nucleic acid molecule or open reading frame, comprising a high proline and alanine content above average is not part of this invention.
  • the invention is based on the fact that a rather large random coil polypeptide or polypeptide segment that does not occur in nature in an isolated manner and that comprises an amino acid sequence consisting solely of proline and alanine residues, wherein said amino acid sequence consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 proline (Pro) and alanine (Ala) residues is provided that is particularly useful in medical/pharmaceutical context.
  • isolated biosynthetic random coil polypeptides or polypeptide segments that do not occur in nature in an isolated manner are also comprised in the herein disclosed (a) biologically active, heterologous protein(s) or (a) protein construct(s) that is/are particularly useful in a pharmaceutical, medical and/or medicinal setting.
  • These biologically active, heterologous proteins/protein constructs comprise as at least one domain of said at least two domains the random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine residues, wherein said amino acid sequence consists of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 proline (Pro) and alanine (Ala) residues.
  • arabinogalactan proteins AGPs
  • Pro-rich proteins Pro-rich proteins
  • extensins belong to a large group of glycoproteins, known as hydroxyproline (Hyp)-rich glycoproteins (HRGPs), which are expressed throughout the plant kingdom.
  • AGP motif comprising an Ala-Pro repeat (AP)51 was expressed as a synthetic glycomodule peptide with N-terminal signal sequence and C-terminal green fluorescent protein in transgenic Arabidopsis thaliana and investigated as a substrate for prolyl hydroxylases and subsequent O-glycosylation of the hydroxyproline residues ( Estévez (2006) Plant Physiol. 142, 458-470 ).
  • the disclosed hydroxylated and/or glycosylated Pro side chains which can form hydrogen bonds to water molecules, appear to have a solubilizing effect.
  • biologically active proteins or protein constructs comprising as (at least) one domain a biosynthetic random coil polypeptide or peptide segment comprising an amino acid sequence consisting solely of proline and alanine residues
  • biosynthetic random coil polypeptides/polypeptide segments described herein are preferably not chemically modified, i.e. they are preferably not glycosylated or hydroxylated.
  • the biosynthetic random coil polypeptide/amino acid sequences of the present invention may comprise concatamers of individual blocks comprising combined proline/alanine stretches of the sequence (Pro) x -(Ala) y , whereby x can have an integer value from 1 to preferably 15, more preferably 1 to 10, even more preferably 1 to 5, and y can have an integer value from 1 to preferably 15, more preferably 1 to 10, even more preferably 1 to5, and x and y can vary between subsequent blocks. Said x and y can also be an integer of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • the amino acid sequences/polypeptides forming random coil conformation in aqueous solution or under physiological conditions may have the formula (I): [Pro x Ala y ] n wherein x is independently selected from integer 1 to 5. Furthermore, for each n, y is independently selected from integer 1 to 5. n, finally, is any integer provided that random coil polypeptide (or segment thereof)/amino acid sequence consists preferably of at least about 50, of at least about 100, of at least about 150, of at least about 200, of at least about 250, of at least about 300, of at least about 350, of at least about 400 amino acid residues and up to about 3000 amino acid residues.
  • polypeptides/amino acid sequences comprising the above concatemers or having the above formula (I) should preferably have the overall length and/or proline/alanine content as defined and explained herein above, i.e. consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 amino acids and/or comprise more than about 10 % and less than about 75 % proline residues.
  • proline/alanine content as defined and explained herein above, i.e. consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 amino acids and/or comprise more than about 10 % and less than about 75 % proline residues.
  • the present invention also relates to random coil polypeptides ((a)polypeptide segment(s))/amino acid sequences comprising an amino acid stretch selected from the group consisting of AAPAAPAPAAPAAPAPAAPA (SEQ ID NO: 1); AAPAAAPAPAAPAAPAPAAP (SEQ ID NO: 2); AAAPAAAPAAAPAAAPAAAP (SEQ ID NO: 3 being an example for [Pro 1 Ala 3 ] 5 ); AAPAAPAAPAAPAAPAAPAAP (SEQ ID NO: 4); APAAAPAPAAAPAPAAAPAPAAAP (SEQ ID NO: 5); AAAPAAPAAPPAAAAPAAPAAPPA (SEQ ID NO: 6) and APAPAPAPAPAPAPAPAPAPAP (SEQ ID NO: 51 being an example for [Pro 1 Ala 1 ] 10 ) or circular permuted versions or (a) multimers(s) of these sequences as a whole or parts of these sequences.
  • the random coil polypeptide ((a) polypeptide segment(s) thereof)/amino acid sequence may comprise the amino acid stretch AAPAAPAPAAPAAPAPAAPA (SEQ ID NO: 1), AAPAAPAPAAPAAPAPAAPA (SEQ ID NO: 1); AAPAAAPAPAAPAAPAPAAP (SEQ ID NO: 2); AAAPAAAPAAAPAAAPAAAP (SEQ ID NO: 3); AAPAAPAAPAAPAAPAAPAAP (SEQ ID NO: 4); APAAAPAPAAAPAPAAAP (SEQ ID NO: 5); AAAPAAPAAPPAAAAPAAPAAPPA (SEQ ID NO: 6) and APAPAPAPAPAPAPAPAP (SEQ ID NO: 51), as well as combinations of these motifs or combinations of fragments and parts of this motifs as long as the resulting biosynthetic random coil polypeptide consists solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala)
  • circular permuted versions of the above amino acid sequences may be used in accordance with the present invention.
  • Exemplary circular permuted versions of e.g. AAPAAPAPAAPAAPAPAAPA can be easily generated, for example by removing the first alanine and adding another alanine at the end of the above sequence.
  • Such a cicular permuted version of SEQ ID NO: 1 would then be APAAPAPAAPAAPAPAAPAA (SEQ ID NO: 7).
  • non-limiting examples of cicular permuted versions of SEQ ID NO. 1 are:
  • Such circular permuted versions may also be considered as examples of a further "module"/"building block” etc. of the herein provided polypeptides/amino acid sequences which can be used accordingly herein.
  • modules and (shorter) fragments or circularly permuted versions of the herein provided amino acid stretches may be used as “modules”, “repeats” and/or building blocks for the herein defined random coil polypeptide (or segment thereof)/amino acid sequence.
  • the random coil polypeptide/amino acid sequence forming random coil conformation may comprise a multimer of any of the above amino acid stretches (or circular permuted versions or fragments thereof), preferably those shown in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6 and SEQ ID NO. 51. It is to note that these sequences are by no means limiting in context of this invention.
  • polypeptides/amino acid sequences comprising the above amino acid stretches (or fragments thereof), circular mutated versions (or fragments thereof) should preferably have the overall length and/or proline/alanine content as defined and explained herein above, i.e. consist of about 50, of about 100, of about 150, of about 200, of about 250, of about 300, of about 350, of about 400 to about 3000 amino acids and/or comprise more than about 10 % and less than about 75 % proline residues. All the definitions given herein above in this context also apply here, mutatis mutandis. Also the term "fragment" has been defined above.
  • the biosynthetic random coil polypeptides (or polypeptide segment)/polymers as provided herein are characterized by a relatively large hydrodynamic volume.
  • This hydrodynamic volume also called apparent size, can easily be determined by analytical gel filtration (also known as size exclusion chromatography, SEC).
  • the random coil polypeptide (or segment thereof) has an apparent size of at least 10 kDa, preferably of at least 25 kDa, more preferably of at least 50 kDa, even more preferably of at least 100 kDa, particularly preferably of at least 200 kDa and most preferably of at least 400 kDa.
  • hydrodynamic volume of specific proteins may include dynamic/static light scattering, analytical ultracentrifugation or analytical gel filtration as exemplified herein below.
  • Analytical gel filtration represents a known method in the art for measuring the hydrodynamic volume of macromolecules.
  • the hydrodynamic volume of a globular polypeptide can be estimated by its molecular weight (Creighton (1993) loc. cit.).
  • the hydrodynamic volume of the polypeptides of the invention consisting preferably of at least about 50, of at least about 100, of at least about 150, of at least about 200, of at least about 250, of at least about 300, of at least about 350, of at least about 400 to about 3000 proline and alanine amino acid residues and having random coil conformation show unexpectedly high values in relation to the hydrodynamic volume that would be estimated for a corresponding folded, globular protein based on the molecular weight.
  • the following relates to biologically active, heterologous proteins or protein constructs comprising, inter alia, the biosynthetic random coil polypeptide (or segment thereof) as described and defined herein above which represent a prefered embodiment of the present invention.
  • biosynthetic random coil polypeptide stretches as provided herein and consisting solely of proline and alanine can, even provide for a higher hydrodynamic volume than a corresponding biosynthetic random coil stretch having the same total number of amino acid residues but consisting solely of proline, alanine and serine (as provided in WO 2008/155134 ).
  • HSA serum albumin
  • Igs immunoglobulins
  • humanized antibodies show long half-lifes, typically of 2 to 3 weeks, which is attributable to their specific interaction with the neonatal Fc receptor (FcRn), leading to endosomal recycling ( Ghetie (2002) Immunol Res, 25:97-113 ).
  • FcRn neonatal Fc receptor
  • most other proteins of pharmaceutical interest in particular recombinant antibody fragments, hormones, interferons, etc. suffer from rapid (blood) clearance. This is particularly true for proteins whose size is below the threshold value for kidney filtration of about 70 kDa ( Caliceti (2003) Adv Drug Deliv Rev 55:1261-1277 ).
  • the recycling mechanism of natural plasma proteins has been employed by producing fusion proteins with the Fc portion of Igs, for example Enbrel®, a hybrid between the extracellular domain of TNF ⁇ receptor and human IgGl ( Goldenberg (1999) Clin Ther 21:75-87 ) or with serum albumin, for example Albuferon® (albinterferon alfa-2b, ZALBINTM, JOULFERON®), a corresponding fusion of IFNalpha with HSA ( Osborn (2002) J Pharmacol Exp Ther 303:540-548 ).
  • Igs for example Enbrel®
  • ZALBINTM ZALBINTM
  • JOULFERON® a fusion of IFNalpha with HSA
  • Albumin with its high plasma concentration of 600 ⁇ M has also been utilized in an indirect manner, serving as carrier vehicle for biopharmaceuticals that are equipped with an albumin-binding function, for example via fusion with a bacterial albumin-binding domain (ABD) from Streptococcal protein G ( Makrides (1996) J Pharmacol Exp Ther 277:534-542 ) or with a peptide selected against HSA from a phage display library ( Dennis (2002) J Biol Chem, 277:35035-35043 ; Nguyen (2006) Protein Eng Des Sel 19:291-297 ).
  • ABS bacterial albumin-binding domain
  • glycosylation analogs of biologically active proteins in which new N-linked glycosylation consensus sequences are introduced has been proposed to prolong plasma half-life; see WO 02/02597 ; Perlman (2003) J Clin Endocrinol Metab 88:2327-2335 ; or Elliott (2003) Nat Biotechnol 21:414-420 ).
  • the described glycoengineered proteins displayed an altered in vivo activity, which indicates that the new carbohydrate side chains influence the biological activity of the engineered protein.
  • the additional carbohydrate side chains are likely to increase the antigenicity of the resulting biological active molecules, which raises substantial safety concerns.
  • fusion proteins comprising the Trypanosoma cruzi derived artificial repetitive sequence PSTAD have been reported to induce a prolonged plasma half-life of trans-sialidase ( Alvarez (2004) JBC 279:3375-3381 ). Yet, such Trypanosoma cruzi derived repeats have been reported to induce a humoral immune response (Alvarez (2004) loc. cit.). Accordingly, alternative strategies to prolong the action of biologically active proteins are desired.
  • biosynthetic amino acid sequences/polypeptides as disclosed herein and consisting solely of proline and alanine according to the invention were surprisingly found to adopt random coil conformation in particular under physiological conditions. Therefore, they are advantageous molecules to provide for the herein below defined "second domain" of the biologically active protein(s)/polypeptide(s), i.e. comprising a polypeptide stretch that forms under physiological conditions a random coil conformation and thereby mediates an increased in vivo and/or in vitro stability to biologically active ("functional") protein(s) or polypeptide(s), in particular, an increased plasma half-life.
  • the hydrodynamic volume of a functional protein that is fused to said random coil domain is dramatically increased as can be estimated by using standard methods mentioned herein.
  • random coil domain Since the random coil domain is thought not to interfere with the biological activity of the first domain of the biologically active protein, the biological activity mediated by the functional protein of interest to which it is fused is essentially preserved.
  • amino acid polymers/polypeptides that form random coil domain as disclosed herein are thought to be biologically largely inert, especially with respect to proteolysis in blood plasma, immunogenicity, isoelectric point/electrostatic behaviour, binding to cell surface receptors as well as internalisation, but still biodegradable, which provides clear advantages over synthetic polymers such as PEG.
  • the present invention relates to a biologically active protein comprising the herein described biosynthetic random coil polypeptide.
  • a biologically active protein/protein construct comprising the biosynthetic random coil polypeptide decribed herein is a heterologous biological active protein/protein construct.
  • herein disclosed is/are also (a) biologically active, heterologous protein(s) comprising or consisting of at least two domains wherein
  • first domain and said “second domain” relate to protein stretches that are not naturally occurring within the same protein or that are not expected to be part of the same hypothetical protein as encoded by a coding nucleic acid sequence (like an open reading frame) as found in nature.
  • said random coil conformation mediates an increased in vivo and/or in vitro stability of said biologically active protein, like the in vivo and/or in vitro stability in biological samples or in physiological environments.
  • proteins comprising a herein defined, additional "second domain” adopting a random coil conformation in aqueous solution or under physiological conditions (for example polymers consisting of about 200 or about 400 or about 600 amino acid residues and comprising PA#1/SEQ ID NO.1, PA#2/SEQ ID NO.2, PA#3/SEQ ID NO.3, PA#4/SEQ ID NO.4, PA#5/SEQ ID NO.5, PA#6/SEQ ID NO. 6 and/or P1A1/SEQ ID NO. 51 as "building blocks) have an advantageous serum stability or plasma half-life, even in vivo, (in particular if intravenously administered) as compared to a control lacking said random coil conformation.
  • polymers consisting of about 200 or about 400 or about 600 amino acid residues and comprising PA#1/SEQ ID NO.1, PA#2/SEQ ID NO.2, PA#3/SEQ ID NO.3, PA#4/SEQ ID NO.4, PA#5/SEQ ID NO.5, PA#6/SEQ ID NO. 6 and/or P
  • WO 2008/155134 (as discussed herein above) it has been shown that biologically active proteins which comprise a domain with an amino acid sequence adopting a random coil conformation have an increased in vivo and/or in vitro stability.
  • the random coil domains disclosed in WO 2008/155134 consist, in particular, of proline, alanine, and serine (PAS) residues. The presence of these three residues is described in this prior art document as an essential requirement for the formation of a stable and soluble random coil in aqueous solution.
  • WO 2007/103515 describes unstructured recombinant polymers which comprise as main constituents a large variety of amino acids, inter alia, glycine, aspartate, alanine, serine, threonine, glutamate and proline.
  • unstructured recombinant polymer has, in contrast to the terms “biosynthetic” and "random coil”, no recognized, clear meaning.
  • WO 2006/081249 protein conjugates comprising a biologically active protein coupled to a polypeptide comprising 2 to 500 units of an amino acid repeat having Gly, Asn, and Gln as a major constituent and Ser, Thr, Asp, Gln, Glu, His, and Asn as a minor constituent. Said protein conjugates are described to have either an increased or a decreased plasma half-life when compared to the unconjugated biologically active protein.
  • WO 2006/081249 does not provide any teaching to predict whether a specific amino acid repeat reduces or augments the plasma half-life of the conjugate.
  • WO 2006/081249 does not teach or suggest that the plasma half-life of proteins can be increased when the conjugated protein comprises an amino acid repeat that forms random coil conformation as shown in the present invention.
  • the amino acid repeats disclosed in WO 2006/081249 comprise at least two residues selected from Gly, Asn, and Gln, which is in clear contrast with the biosynthetic random coil polypeptides of the present invention which comprise an amino acid sequence that solely consists of proline and alanine amino acid residues.
  • biosynthetic random coil amino acid sequences as provided herein which, in contrast to the prior art, solely comprise proline and alanine residues (i.e. which preferably do not comprise a substantial amount of any other amino acid, also not a substantial amount of serine or no serine at all) do also form a useful random coil structure.
  • proline and alanine residues i.e. which preferably do not comprise a substantial amount of any other amino acid, also not a substantial amount of serine or no serine at all
  • SA serine and alanine
  • biological activity describes the biological effect of a substance on living matter.
  • biologically active protein as used herein relate to proteins that are capable of inducing a biological effect in living cells/organisms that are exposed to said protein or polypeptide.
  • biologically active protein relates to the whole protein of the invention which both comprises an amino acid sequence having and/or mediating said biological activity (said first domain) and the inventive amino acid sequence adopting/forming random coil conformation and consisting solely of proline and alanine (said second domain).
  • amino acid sequence having and/or mediating biological activity or “amino acid sequence with biological activity” as used herein to the above-defined “first domain” of the biologically active protein of the invention, which mediates or has or is capable of mediating or having the above defined “biological activity”.
  • amino acid sequence having and/or mediating biological activity or “amino acid sequence with biological activity” are any proteins of interest (and functional fragments thereof, such as antibody fragments, fragments comprising extracellular or intracellular domain(s) of a membrane receptor, truncated forms of a growth factor or cytokine and the like), the half-life of which, either in vivo or in vitro, needs to be prolonged.
  • amino acid sequence having and/or mediating biological activity in accordance with the present invention may be deduced from any "protein of interest", i.e. any protein of pharmaceutical or biological interest or any protein that is useful as a therapeutic/diagnostic agent.
  • the biologically active proteins may comprise a first domain comprising a biologically active amino acid sequence which is derived from naturally produced polypeptides or polypeptides produced by recombinant DNA technology.
  • the protein of interest may be selected from the group consisting of binding proteins/binding molecules, immunoglobulins, antibody fragments, transport proteins, membrane receptors, signaling proteins/peptides such as cytokines, growth factors, hormones or enzymes and the like.
  • the random coil polypeptide (or polypeptide segment) comprised in the second domain of the biologically active protein forms the random coil conformation in particular under physiological conditions. This is particularly relevant in context of biologically active proteins which may form part of a pharmaceutical composition that is to be administered to a subject or patient.
  • the inventive biosynthetic random coil domain (said "second domain”) of the biologically active protein natively adopts/forms/has random coil conformation, in particular in vivo and when administered to mammals or human patients in need of medical intervention.
  • proteins having a non-random secondary and/or tertiary structure as native conformation tend to adopt a random coil conformation under non-physiological conditions (i.e. under denaturing conditions).
  • denatured proteins have completely different characteristics compared to the biologically active protein comprising the random coil polypeptide of the present invention.
  • the "biologically active protein” and the biologically active part of the fusion proteins/fusion constructs as provided herein maintain their biological function also when combined and/or linked with the biosynthetic random coil polypeptide (or polypeptide segment) of this invention.
  • the random coil polypeptide retains solubility under physiological conditions.
  • the protein construct of the present invention may comprise the "second", random coil forming/adopting domain transiently or temporarily not in random coil conformation, for example, when in form of a specific composition, like a lyophylisate or dried composition.
  • a “second domain” of the inventive protein construct again adopts after, e.g., reconstitution in corresponding buffers (preferably “physiological” buffers/excipients and/or solvents), the herein defined random coil conformation.
  • Said "second domain” is (if necessary, after corresponding reconstitution) capable of mediating an increased in vivo and/or in vitro stability of the inventive biologically active protein. It is preferred herein that the "second domain” as defined herein consists of the random coil polypeptide (or polypeptide segment) of the present invention.
  • domain relates to any region/part of an amino acid sequence that is capable of autonomously adopting a specific structure and/or function.
  • a domain may represent a functional domain or a structural domain.
  • the proteins of the present invention comprise at least one domain/part having and/or mediating biological activity and at least one domain/part forming random coil conformation.
  • the proteins of the invention also may consist of more than two domains and may comprise e.g. an additional linker or spacer structure between the herein defined two domains/parts or another domain/part like, e.g.
  • a protease sensitive cleavage site an affinity tag such as the His 6 -tag or the Strep -tag , a signal peptide, retention peptide, a targeting peptide like a membrane translocation peptide or additional effector domains like antibody fragments for tumour targeting associated with an anti-tumour toxin or an enzyme for prodrug-activation etc.
  • the biologically active protein of the invention has a hydrodynamic volume as determined by analytical gel filtration (also known as size exclusion chromatography, SEC) of at least 50 kDa, preferably of at least 70 kDa, more preferably of at least 80 kDa, even more preferably of at least 100 kDa, particularly preferably of at least 125 kDa and most preferably of at least 150 kDa.
  • analytical gel filtration also known as size exclusion chromatography, SEC
  • SEC size exclusion chromatography
  • the hydrodynamic volume of the biologically active proteins of the invention that comprise the above defined second domain i.e. the domain comprising or consisting of the herein provided random coil polypeptide (or segment thereof) are shown to have an unexpectedly large hydrodynamic volume in relation to the estimated hydrodynamic volume for a corresponding folded, globular protein based on their molecular weight or number/composition of amino acid residues.
  • the first domain comprising an "amino acid sequence having and/or mediating biological activity” may also adopt its biological activity in the context of or after association with another polypeptide or amino acid sequence.
  • the Fab fragment of an antibody such as the one of the anti-tumour antibody Herceptin ( Eigenbrot (1993) J. Mol. Biol. 229:969-995 ) consists of two different polypeptide chain, the immunoglobulin light chain and a fragment of the immunoglobulin heavy chain, which may furthermore be linked via (an) interchain disulfide bond(s). According to the present invention, it may be sufficient to link one of those chains (e.g.
  • reconstitution may be achieved, for example, by co-expression of the different polypeptides (on the one hand a fusion protein of one chain and the random coil polypeptide, on the other hand the other chain) in the same host cell, as described in the appended examples, or by reconstitution in vitro, for example, as part of a refolding protocol.
  • the first domain as defined herein may comprise two separate polypeptide chains which are linked only non-covalently.
  • the independent chains of the biologically active protein/domain may each be linked to the random coil polypeptide (or polypeptide segment).
  • binding protein relates to a molecule that is able to specifically interact with (a) potential binding partner(s) so that it is able to discriminate between said potential binding partner(s) and a plurality of different molecules as said potential binding partner(s) to such an extent that, from a pool of said plurality of different molecules as potential binding partner(s), only said potential binding partner(s) is/are bound, or is/are significantly bound.
  • Methods for the measurement of binding between a binding protein and a potential binding partner are known in the art and can be routinely performed, e.g., by using ELISA, isothermal titration calorimetry, equilibrium dialysis, pull down assays, surface plasmon resonance or a Biacore apparatus.
  • binding proteins/binding molecules which are useful in the context of the present invention include, but are not limited to antibodies, antibody fragments such as Fab fragments, F(ab') 2 fragments, single chain variable fragments (scFv), isolated variable regions of antibodies (VL and/or VH regions), CDRs, single domain antibodies/immunoglobulins, CDR-derived peptidomimetics, lectins, immunoglobulin domains, fibronectin domains, protein A domains, SH3 domains, ankyrin repeat domains, lipocalins or various types of scaffold-derived binding proteins as described, for example, in Skerra (2000) J Mol Recognit 13:167-187 , Gebauer (2009) Curr Opin Chem Biol 13:245-255 , Binz (2005) Nat Biotechnol 23:1257-1268 or Nelson (2009) Nat Biotechnol 27:331-337 .
  • exemplary biologically active proteins of interest include, but are not limited to, granulocyte colony stimulating factor, human growth hormone, alpha-interferon, beta-interferon, gamma -interferon, lambda-interferone, tumor necrosis factor, erythropoietin, coagulation factors such as coagulation factor VIII, coagulation factor VIIa, coagulation factor IX, gp120/gp160, soluble tumor necrosis factor I and II receptor, thrombolytics such as reteplase, peptides with metabolic effects such as GLP-1 or exendin-4, immunosuppressive/immunoregulatory proteins like interleukin-1 receptor antagonists or anakinra, interleukin-2 and neutrophil gelatinase-associated lipocalin or other natural or engineered lipocalins or those proteins or compounds listed
  • biologically active proteins in particular proteins comprised in the first domain or constituting/being the first domain of the biologically active protein
  • proteins which may be employed in context of the present invention are, inter alia, follicle-stimulating hormone, glucocerebrosidase, thymosin alpha 1, glucagon, somatostatin, adenosine deaminase, interleukin 11, hematide, leptin, interleukin-20, interleukin-22 receptor subunit alpha (IL-22ra), interleukin-22, hyaluronidase, fibroblast growth factor 18, fibroblast growth factor 21, glucagon-like peptide 1, osteoprotegerin, IL-18 binding protein, growth hormone releasing factor, soluble TACI receptor, thrombospondin-1, soluble VEGF receptor Flt-1, ⁇ -galactosidase A, myostatin antagonist, gastric inhibitory polypeptide, alpha-1 antitrypsin, IL-4
  • the present invention also relates to comprising the biosynthetic random coil proline/alanine polypeptide or proline/alanine polypeptide segment and pharmaceutically or medically useful molecules, like small molecules, peptides or biomacromolecules such as proteins, nucleic acids, carbohydrates, lipid vesicles and the like, in particular pharmaceutically or medically useful proteins, like (but not limited to) binding proteins/binding molecules, immunoglobulins, antibody fragments, transport proteins, membrane receptors, signaling proteins/peptides, cytokines, growth factors, hormones or enzymes and the like may be comprised in the herein defined drug constructs btu they may also be part of the herein defined biologically active, heterologous protein comprising or consisting of said defined at least two domains.
  • pharmaceutically or medically useful molecules like small molecules, peptides or biomacromolecules such as proteins, nucleic acids, carbohydrates, lipid vesicles and the like, in particular pharmaceutically or medically useful proteins, like (
  • said particular pharmaceutically or medically useful proteins may be the "first domain" of said at least two domains comprising or consisting of an amino acid sequence having and/or mediating said biological activity
  • Functional fragments are fragments of said pharmaceutically or medically useful proteins that are still capable to elucidate the desired biological or pharmaceutical response in vivo and/or in vitro and/or still have or mediate the desired biological activity.
  • polypeptide linker/spacer inserted between said first and said second domains, preferably comprises plural hydrophilic, peptide-bonded amino acids that are covalently linked to both domains.
  • said polypeptide linker/spacer comprises a plasma protease cleavage site which allows the controlled release of said first domain comprising a polypeptide having and/or mediating a biological activity. Linkers of different types or lengths may be identified without undue burden to obtain optimal biological activity of specific proteins.
  • the biologically active proteins of the present invention are fusion proteins.
  • a fusion protein as described herein may comprise at least one domain which can mediate a biological activity and at least one other domain which comprises the biosynthetic random coil polypeptide (or polypeptide segment) as described herein in a single multi-domain polypeptide.
  • the present invention is not limited to fusion proteins wherein one domain mediates a biological activity.
  • other "fusion proteins"/"fusion constructs" are provided herein wherein one part/domain is or comprises the inventive random coil polypeptide/polymer of proline/alanine and the other part/domain comprises another protein stretch/structure.
  • the random coil polypeptide (or polypeptide segment) does not necessarily carry Pro or Ala residues at its amino or carboxyl terminus.
  • the biologically active protein in accordance with the present invention may represent a protein conjugate wherein a protein of interest or a polypeptide/polypeptide stretch/peptide/amino acid sequence having and/or mediating biological activity is conjugated via a non-peptide bond to an amino acid sequence which forms/adopts random coil conformation, in particular, the random coil polypeptide (or polypeptide segment) as provided herein and consisting solely of proline and alanine residues.
  • Non-peptide bonds that are useful for cross-linking proteins are known in the art with the biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues as provided herein.
  • Such Non-peptide bonds may include disulfide bonds, e.g.
  • Cys side chains thioether bonds or non-peptide covalent bonds induced by chemical cross-linkers, such as disuccinimidyl suberate (DSS) or sulfosuccinimidyl 4-[p-maleimidophenyl] butyrate (Sulfo-SMPB), metal-chelating/complexing groups, as well as non-covalent protein-protein interactions.
  • chemical cross-linkers such as disuccinimidyl suberate (DSS) or sulfosuccinimidyl 4-[p-maleimidophenyl] butyrate (Sulfo-SMPB), metal-chelating/complexing groups, as well as non-covalent protein-protein interactions.
  • the "biologically active protein" of the present invention may also comprise more than one "amino acid sequence having and/or mediating a biological activity". Furthermore, the biologically active protein may also comprise more than biosynthetic random coil polypeptide (or segment thereof). In the simplest case, the biologically active protein consists of two domains, i.e. a first domain comprising an amino acid sequence having and/or mediating a biological activity and a second domain comprising the biosynthetic polypeptide (or segment thereof).
  • the present invention is not limited to "biologically or therapeutically active proteins" linked to the herein disclosed biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues.
  • other proteins or molecules of interest as relevant for other industries, like food or beverage industry, cosmetic industry and the like, may be manufactured by the means and methods provided herein.
  • domain comprising an amino acid sequence having and/or mediating a biological activity and the “second domain comprising the random coil polypeptide (or segment thereof) as comprised in the biologically active proteins of the invention may be organized in a specific order.
  • first and second domain of the inventive biologically active polypeptide may be arranged in an order, whereby said "first domain” (i.e. protein of interest; “amino acid sequence having and/or mediating said biological activity") is located at the amino (N-) terminus and said "second domain” (i.e. the domain that comprises the herein provided random coil polypeptide (or segment thereof)) is located at the carboxy (C-) terminus of the biologically active protein.
  • first domain i.e. protein of interest; "amino acid sequence having and/or mediating said biological activity
  • second domain i.e. the domain that comprises the herein provided random coil polypeptide (or segment thereof)
  • amino acid sequence having and/or mediating said biological activity is located at the carboxy (C-) terminus and said "second domain” (i.e. the domain that comprises the herein provided random coil polypeptide (or segment thereof)) is located at the amino (N-) terminus of the biologically active protein.
  • the domain order may, accordingly, be (from N-terminus to C-terminus): first domain (amino acid sequence having and/or mediating a biological activity) - second domain (random coil polypeptide (or segment thereof)).
  • the domain order may be (from N-terminus to C-terminus): second domain (random coil polypeptide (or segment thereof)) - first domain (amino acid sequence having and/or mediating a biological activity).
  • the biologically active protein may comprise two "first domain", i.e. two specific amino acid sequences having and/or mediating a biological activity, whereby this biological activity may be the same or a different activity.
  • the domain order may be (from N-terminus to C-terminus): first domain (amino acid sequence having and/or mediating a specific biological activity) - second domain (random coil polypeptide (or segment thereof)) - first domain (amino acid sequence having and/or mediating a specific (optionally different) biological activity).
  • the biologically active protein comprises more than one "second domain” (i.e. the biologically active protein comprises more than one random coil polypeptide (or segment thereof).
  • the biologically active protein consists of two such "second domains", i.e two domains comprising the biosynthetic random coil polypeptide (or segment thereof), and one "first domain” (comprising an amino acid sequence having and/or mediating a biological activity)
  • the domain order may be (from N-terminus to C-terminus): second domain (random coil polypeptide (or segment thereof)) - first domain (amino acid sequence having and/or mediating a specific biological activity) -- second domain (random coil polypeptide (or segment thereof)).
  • the biologically active protein comprises more than one "second domain” it is envisaged herein that these "second domains" may be identical or may be different.
  • the biologically active protein may comprise more than one "first domain", i.e. more than one specific amino acid sequences having and/or mediating a biological activity and more than one "second domain” (biosynthetic random coil polypeptide (or segment thereof)) whereby these "first domains" may be identical or different and/or whereby said "second domains” may be identical or different.
  • first domain i.e. more than one specific amino acid sequences having and/or mediating a biological activity
  • second domain biosynthetic random coil polypeptide (or segment thereof)
  • said domain(s) comprising an amino acid sequence having and/or mediating the said biological activity may also be a biologically active fragment of a given protein with a desired biological function. Therefore, the herein defined "second domain" (preferably comprising the herein provided random coil polypeptide (or segment thereof)) may also be located between two biologically active fragments of a protein of interest or between biologically active fragments of two proteins of interest. All the explanations and definitions given herein above in context of "full length" proteins/polypeptides of interest (i.e. when the amino acid sequences has/mediates a certain biological activity on its own) apply, mutatis mutandis, in context of such fragments.
  • the above invention is not limited to the constructs that comprise a "domain” with a “biological active function”.
  • the constructs of the present invention may also comprise domains with other functions and are not limited to biological activities. These are merely embodiments of the present invention and it is evident for the skilled artisan that other constructs can easly be made and used without deferring from the gist of the present invention. Accordingyl, the herein said in context of "amino acid sequence having and/or mediating a specific biological activity" applies, mutatis mutnatis, for other constructs, for example constructs to be used in other technical fields, like in cosmetics, food processing, dairy products, paper production, etc. As mentioned herein above, the biosynthetic polypeptides/polymers of the rpesent invention can also be used to be linked with e.g. small molecules and the like.
  • amino acid sequence having and/or mediating first biological activity is not limited to full-length polypeptides that have and/or mediate said biological activity or function, but also to biologically and/or pharmacologically active fragments thereof.
  • first domains as defined herein are comprised in the inventive "biologically active protein” it is also envisaged that these "first domains" are or represent different parts of a protein complex or fragments of such parts of protein complex.
  • the biologically active proteins of the invention which are modified to comprise a random coil polypeptide surprisingly exhibit an increased in vivo and/or in vitro stability when compared to unmodified biologically active proteins that lack said random coil domain.
  • in vivo stability relates to the capacity of a specific substance that is administered to the living body to remain biologically available and biologically active. In vivo, a substance may be removed and/or inactivated due to excretion, kidney filtration, liver uptake, aggregation, degradation and/or other metabolic processes.
  • biologically active proteins that have an increased in vivo stability may be less rapidly excreted through the kidneys (urine) or via the feces and/or may be more stable against proteolysis, in particular against in vivo proteolysis in biological fluids, like blood, liquor cerebrospinalis, peritoneal fluid, and lymph.
  • the increased in vivo stability of a biologically active protein manifests in a prolonged plasma half-life of said biologically active protein.
  • the increased in vivo stability of the biologically active protein is a prolonged plasma half-life of said biologically active protein comprising said second domain when compared to the biologically active protein lacking the second domain.
  • biologically active proteins may be specifically detected in the blood plasma using Western blotting techniques or enzyme linked immunosorbent assay (ELISA).
  • ELISA enzyme linked immunosorbent assay
  • other methods may be employed to specifically measure the plasma half-life of a protein of interest. Such methods include, but are not limited to the physical detection of a radioactively labelled protein of interest. Methods for radioactive labelling of proteins e.g. by radioiodination are known in the art.
  • the term "increased in vitro stability" as used herein relates to the capacity of a biologically active protein to resist degradation and/or aggregation and to maintain its original biological activity in an in vitro environment. Methods for measuring the biological activity of biologically active proteins are well known in the art.
  • a drug conjugate which comprises the herein described and defined random coil polypeptide or polypeptide segment and a small molecule drug that is conjugated to said random coil polypeptide or polypeptide segment.
  • the small molecules are doxorubicin, calicheamicin, camptothecin, fumagillin, dexamethasone, geldanamycin, paclitaxel, docetaxel, irinotecan, cyclosporine, buprenorphine, naltrexone, naloxone, vindesine, vancomycin, risperidone, aripiprazole, palonosetron, granisetron, cytarabine, NX1838, leuprolide, goserelin, buserelin, octreotide, teduglutide, cilengitide, abarelix, enfuvirtide, ghrelin and derivatives, tubulysins, platin
  • DNA or RNA aptamers, peptides, peptidomimetics also relates to drug constructs comprising the herein defined random coil polypeptide or polypeptide segment and in particular pharmaceutically or medically useful molecules, like small molecules, peptides or biomacromolecules such as proteins, nucleic acids, carbohydrates, lipid vesicles and the like.
  • pharmaceutically or medically useful molecules like small molecules, peptides or biomacromolecules such as proteins, nucleic acids, carbohydrates, lipid vesicles and the like.
  • the present Figures and experimental information in the corresponding figure legends provide for illustrative examples, wherein the herein disclosed drug conjugates comprise (i) a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues, and (ii) a small molecule, which is, as illustration, selected from digoxigenin and fluorescein. It is of note that these are not only academic examples.
  • Fluorescein or fluorescein derivates are commonly used as diagnostics, and medical fluorescein solutions are sold under the trade names Fluoescite®, AK-FLUOR® or Fluress®. Such compounds can certainly profit from the means and methods provided herein.
  • Digoxigenin forms the steroid part of digoxin, a well known secondary plant metabolite with cardioactive function which furthermore contains three digitoxose sugars.
  • Digoxin, and to a lesser extent the closely related compound digitoxin are widely used for the treatment of ventricular tachyarrhythmias and congestive heart failure ( Hauptman (1999) Circulation 99: 1265-1270 ). All cardioactive steroids are potent and highly specific inhibitors of the Na + /K + -ATPase located in the cellular plasma membrane, thereby exerting sympatholytic or positive inotropic effects.
  • the amino acid polymer forming random coil conformation / the random coil polypeptide (or segment thereof) as defined and provided herein can be conjugated to a small molecule/small molecule drug.
  • plasma half- life and/or solubility of the small molecule/small molecule drug may be increased, unspecific toxicity may be decreased, and the prolonged exposure of active drug to target cells or structures in the body may result in enhanced pharmacodynamics.
  • N-terminus of the random coil polypeptide with an activated drug derivative, e.g. as N-hydroxysuccinimide (NHS) ester derivative ( Hermanson (1996) Bioconjugate Techniques, Academic Press, San Diego, CA ), is possible.
  • an activated drug derivative e.g. as N-hydroxysuccinimide (NHS) ester derivative ( Hermanson (1996) Bioconjugate Techniques, Academic Press, San Diego, CA ).
  • the N-terminal amino group can be chemically coupled with a wide variety of functional groups such as aldehydes and ketones (to form Schiff bases, which may be reduced to amines using sodium borohydride or sodium cyanoborohydride, for example) or to activated carbonic acid derivatives (anhydrides, chlorides, esters and the like, to form amides) or to other reactive chemicals such as isocyanates, isothiocyanates, sulfonly chlorides etc.
  • the N-terminus of the amino acid polymer/polypeptide can first be modified with a suitable protective group, for example an acetyl group, a BOC group or an FMOC group ( Jakubke (1996) Peptide.
  • the amino terminus may be protected by a pyroglutamyl group, which can form from an encoded Gln amino acid residue preceding the Pro/Ala polypeptide or polypeptide segment.
  • a pyroglutamyl group which can form from an encoded Gln amino acid residue preceding the Pro/Ala polypeptide or polypeptide segment.
  • the N-terminus or the C-terminus of the amino acid polymer forming random coil conformation / the random coil polypeptide can be modified with a commercially available linker reagent providing a maleimide group, thus allowing chemical coupling to a thiol group as part of the drug molecule. In this manner uniform drug conjugates can be easily obtained. Similar techniques, which are well known in the art (Hermanson (1996) loc. cit.), can be used to couple the random coil polypeptide to a peptide or even to a protein drug. Such peptides or proteins can easily be prepared carrying a Lys or Cys side chain, which allows their in vitro coupling to the amino acid polymer forming random coil conformation via NHS ester or maleimide active groups.
  • similar drug conjugates can be prepared with fusion proteins comprising the random coil polypeptide (or segment thereof). Yet, and as illustrated in the appended Examples and Figures the present invention also provides for the preparation of a random coil polypeptides or a random coil polypeptide segments as comprised in the innovative conjugates of the present invention.
  • the random coil polypeptide may be equipped with additional side chains, at the N- or C-terminus or internally, suitable for chemical modification such as lysine residues with their ⁇ -amino groups, cysteine residues with their thiol groups, or even non-natural amino acids, allowing the conjugation of multiple small molecules using, for example, NHS ester or maleimide active groups.
  • a prodrug may be linked transiently to the random coil polypeptide.
  • the linkage can be designed to be cleaved in vivo, in a predictable fashion, either via an enzymatic mechanism or by slow hydrolysis initiated at physiological pH similarly as, for example, the poorly soluble antitumor agent camptothecin was conjugated to a PEG polymer, thus achieving increased biodistribution, decreased toxicity, enhanced efficacy and tumor accumulation ( Conover (1998) Cancer Chemother Pharmacol, 42:407-414 ).
  • Examples for further prodrugs are chemotherapeutic agents like docetaxel ( Liu (2008) J Pharm Sci. 97:3274-3290 ), doxorubicin ( Veronese (2005) Bioconjugate Chem. 16: 775-784 ) or paclitaxel ( Greenwald (2001) J Control Release 74:159-171 ).
  • the small molecule may be coupled to a fusion protein comprising the amino acid polymer/polypeptide genetically fused to a targeting domain, e.g. an antibody fragment, thus resulting in a specific delivery of the small molecule drug.
  • a targeting domain e.g. an antibody fragment
  • immunotoxins can be easily generated by conjugation with a cytotoxic small molecule if the targeting domain is directed against a cell-surface receptor which undergoes internalization, for example.
  • the present invention also relates, therefore, to the provision of the herein disclosed biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues for further and additional coupling with other compounds of choice.
  • Said further and/or additional coupling may be and/or may comprise the first coupling of said biosynthetic random coil polypeptide or biosynthetic random coil polypeptide segment with or to another compound.
  • nucleic acid molecules encoding the random coil polypeptides (or segments thereof) or biologically active proteins as described herein.
  • said nucleic acid molecule may comprise a nucleic acid sequence encoding a polypeptide having biological activity and a nucleic acid sequence encoding the random coil polypeptide (or segment thereof)..
  • nucleic acid molecule is intended to include nucleic acid molecules such as DNA molecules and RNA molecules. Said nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • said nucleic acid molecule may be comprised in a vector.
  • the present disclosure also relates to a nucleic acid molecule encoding the random coil polypeptide or polypeptide segment as comprised in the conjugates provided herein, like a drug conjugate as defined herein.
  • the present invention relates to a nucleic acid molecule encoding a protein conjugate that comprises a biologically active protein as defined above and that comprises, additionally, a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues.
  • nucleic acid molecule that encodes a drug conjugate as defined above, said nucleic acid molecule comprising
  • the above mentioned "translated amino acid and/or a leader sequence" under (i) may for example be the starting "M", i.e. a methionine derived from a corresponding starting codon, it may also comprise non-translated sequences of an mRNA like the 5' sequence up to a start codon which comprises for example a ribosome binding site. Such a sequence may however also comprise classical leader and/or signal sequences for example for secretion of an expressed protein into the periplasm or in a culture medium.
  • Prokaryotic signal peptides are for example OmpA, MalE, PhoA, DsbA, pelB, Afa, npr, STII.
  • Eukaryotic signal peptides are for example Honeybee melittin signal sequence, acidic glycoprotein gp67 signal sequence, mouse IgM signal sequence, hGH signal sequence.
  • Biologically active proteins or polypeptides that comprises or that is an amino acid sequence that has or that mediates a biological activity as well as other proteins of interest, like a protein to be employed in other industrial areas, have been provided herein above. Said embodiments apply, mutatis mutantis, for the nucleic acid molece (part/segments (iii)) as illustrated herein above.
  • Translational stop codons to be employed in the nucleic acid molecule provided herein are well known in the art and are, e.g. codons UAA, UAG or UGA.
  • nucleic acid molecule as provided herein above said nucleic acid molecule parts/segments (ii) and (iii) are interchanged in their position on said nucleic acid molecule encoding for a drug conjugate.
  • nucleic acid molecule would comprise the following order of parts/segments:
  • the nucleic acid molecules as provided herein above may also, optionally, comprise, between parts/segments (i) and (ii) and/or between parts/segments (ii) and (iii), a protease and/or a chemical cleavage site and/or a recognition site.
  • chemical cleavage sites are well known in the art, and comprise for example specific, individual amino acid sequences (see, e.g. Lottspeich and Engels (Hrsg.) (2006) Bioanalytik. 2. Auflage. Spektrum Akademischer Verlag, Elsevier, Ober, Germany ).
  • cyanogen bromide or cyanogen chloride cleaves the peptide bond following a Met residue; hydroxylamine cleaves the asparaginyl-glycyl bond; formic acid cleaves Asp-Pro; 2-(2'-nitrophenylsulfenyl)-3-methyl-3-bromoinolenine, 2-iodosobenzoic acid or N-chlorosuccinimide after Trp; 2-nitro-5-thiocyanatobenzoic acid after Cys.
  • residue preceding the Pro/Ala polypeptide or polypeptide segment may be substituted to Met via site-directed mutagenesis and the resulting fusion protein can then be cleaved by BrCN.
  • other amino acid sequences comprising cleavage site can be introduced into the recombinant fusion protein or its encoding nucleic acid by way of site-directed mutagenesis.
  • protease recognition/cleavage sites comprise, but are not limited to: trypsin, chymotrypsin, enterokinase, Tobacco Etch Virus (TEV) protease, PreScission protease, HRV 3C Protease, SUMO Protease, Sortase A, granzyme B, furin, thrombin, factor Xa or self cleavable inteins.
  • TEV Tobacco Etch Virus
  • SUMO Protease SUMO Protease
  • Sortase A granzyme B
  • furin furin
  • thrombin factor Xa or self cleavable inteins.
  • Factor Xa hydrolyses the peptide bond at the C-terminal end of the amino acid sequence IleGluGlyArg, which may be inserted between the N-terminal fusion partner and the Pro/Ala polypeptide or polypeptide segment.
  • a particularly simple method to achieve proteolytic cleavage would be by insertion or substitution of a Lys or Arg side chain at the N-terminal end of the Pro/Ala polypeptide or polypeptide segment followed by digest with trypsin, which does not cleave within the Pro/Ala polypeptide or polypeptide segment as long as internal Lys or Arg side chains are avoided.
  • Illustrative recognition sites are, without being limited, D-D-D-D-K (enterokinase), ENLYFQ(G/S) (TEV protease), I-(E/D)-G-R (Factor Xa), L-E-V-L-F-Q-G-P (HRV 3C), R-X-(K/R)-R (Furin), LPXTG (Sortase A), L-V-P-R-G (Thrombin) or I-E-X-D-X-G (Granzyme B).
  • the present invention provides for recombinantly produced biosynthetic random coil polypeptides and polypeptide segments that can be conjugated with pharmaceutically active polypeptides or small molecules.
  • nucleic acid encoding for a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues, said nucleic acid molecule comprising
  • Such a nucleic acid molecule may, optionally, comprise, between (i) and (ii) a protease and/or a chemical cleavage site and/or a recognition site.
  • nucleic acid molecule i.e. a protease and/or a chemical cleavage site and/or a recognition
  • embodiments provided herein above in context of the first two described nucleic acid molecules i.e. a protease and/or a chemical cleavage site and/or a recognition
  • Useful and illustrative signal sequences to be employed in context of this invention comprise, but are not limited, prokaryotic sequences like: OmpA, MalE, PhoA, DsbA, pelB, Afa, npr, STII or eukaryotic sequences like: Honeybee melittin signal sequence, acidic glycoprotein gp67 signal sequence, mouse IgM signal sequence, hGH signal sequence.
  • nucleic acid molecule encoding the biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues of the present invention is useful in methods as also provided herein below and as illustrated in the appended examples and figures.
  • Such an expressed random coil polypeptide or random coil polypeptide segment can be isolated from, e.g. host cells expressing such a random coil polypeptide or random coil polypeptide segment.
  • host cells may be transfected cells, for example with an vector as provided herein.
  • the present invention relates to nucleic acid molecules which upon expression encode the random coil polypeptide (or segment thereof) or biologically active proteins of the invention. Yet, in a further embodiment, the present invention relates to nucleic acid molecules which upon expression encode the herein disclosed polypeptides that, entirely or in part, form/adopt random coil conformation in aqueous solution or under physiological conditions. Said nucleic acid molecules may be fused to suitable expression control sequences known in the art to ensure proper transcription and translation of the polypeptide as well as signal sequences to ensure cellular secretion or targeting to organelles. Such vectors may comprise further genes such as marker genes which allow for the selection of said vector in a suitable host cell and under suitable conditions.
  • the nucleic acid molecule of the invention is comprised in a recombinant vector in which a nucleic acid molecule encoding the herein described biologically active protein is operatively linked to expression control sequences allowing expression in prokaryotic or eukaryotic cells.
  • Expression of said nucleic acid molecule comprises transcription of the nucleic acid molecule into a translatable mRNA.
  • Regulatory elements permitting expression in prokaryotic host cells comprise, e.g., the lambda PL, lac, trp, tac, ara, phoA, tet or T7 promoters in E. coli.
  • Possible regulatory elements ensuring expression in eukaryotic cells are well known to those skilled in the art. They usually comprise regulatory sequences ensuring initiation of transcription and optionally poly-A signals effecting termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers, and/or naturally associated or heterologous promoter regions. Examples for regulatory elements permitting expression in eukaryotic host cells are the AOX1 or GAL1 promoters in yeast or the CMV, SV40, RSV (Rous sarcoma virus) promoters, CMV enhancer, SV40 enhancer or a globin intron in mammalian and other animal cells. Apart from elements which are responsible for the initiation of transcription such regulatory elements may also comprise transcription termination signals, such as the SV40-poly-A site or the tk-poly-A site, downstream of the coding region.
  • the present invention also relates to vectors, particularly plasmids, cosmids, viruses, and bacteriophages that are conventionally employed in genetic engineering, comprising a nucleic acid molecule encoding the the random coil polypeptide (or segment thereof) or the biologically active protein of the invention.
  • said vector is an expression vector and/or a gene transfer or targeting vector.
  • Expression vectors derived from viruses such as retroviruses, vaccinia virus, adeno-associated virus, herpes viruses or bovine papilloma virus may be used for delivery of the polynucleotides or vector of the invention into targeted cell populations.
  • the vectors containing the nucleic acid molecules of the invention can be transfected into the host cell by well known methods, which vary depending on the type of cell. Accordingly, the invention further relates to a cell comprising said nucleic acid molecule or said vector. Such methods, for example, include the techniques described in Sambrook (1989), loc. cit. and Ausubel (1989), loc. cit. Accordingly, calcium chloride transfection or electroporation is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or electroporation may be used for other cellular hosts (Sambrook (1989), loc. cit.). As a further alternative, the nucleic acid molecules and vectors of the invention can be reconstituted into liposomes for delivery to target cells.
  • the nucleic acid molecule or vector of the invention which is present in the host cell may either be integrated into the genome of the host cell or it may be maintained extra-chromosomally. Accordingly, the present invention also relates to a host cell comprising the nucleic acid molecule and/or the vector of this invention.
  • Host cells for the expression of polypeptides are well known in the art and comprise prokaryotic cells as well as eukaryotic cells, e.g. E.
  • coli cells coli cells, yeast cells, invertebrate cells, CHO cells, CHO-K1 cells, HEK 293 cells, Hela cells, COS-1 monkey cells, melanoma cells such as Bowes cells, mouse L-929 cells, 3T3 cell lines derived from Swiss, Balb-c or NIH mice, BHK or HaK hamster cell lines and the like.
  • the present invention comprises methods for the preparation of the conjugates of the present invention as well as the biosynthetic random coil polypeptide (or segment thereof) or biologically active proteins provided herein and comprising culturing the (host) cell of this invention and isolating said random coil polypeptide (or segment thereof) or the conjugate or a biologically active protein from the culture as described herein.
  • inventive random coil polypeptide (or segment thereof), the conjugate or biologically active protein comprising a random coil domain may be produced by recombinant DNA technology, e.g.
  • inventive biologically active protein or random coil polypeptide may be produced in any suitable cell culture system including prokaryotic cells, e.g. E. coli BL21, KS272 or JM83, or eukaryotic cells, e.g. Pichia pastoris, yeast strain X-33 or CHO cells. Further suitable cell lines known in the art are obtainable from cell line depositories like the American Type Culture Collection (ATCC).
  • ATCC American Type Culture Collection
  • prokaryotic is meant to include bacterial cells while the term “eukaryotic” is meant to include yeast, higher plant, insect and mammalian cells.
  • the transformed hosts can be grown in fermentors and cultured according to techniques known in the art to achieve optimal cell growth.
  • the present invention relates to a process for the preparation of a random coil polypeptide (or segment thereof) or a biologically active protein described above comprising cultivating a cell of the invention under conditions suitable for expression of the biologically active protein or random coil polypeptide (or segment thereof) and isolating said protein/polypeptide from the cell or the culture medium.
  • the random coil polypeptide (or segment thereof) per se of the present invention does, preferably not comprises any chemically reactive group, except for, possibly, one N-terminal primary (or, in the case of proline, secondary) amino group and one carboxylate group at the C-terminus of the polymer.
  • the biosynthetic random coil polypeptide/polymer as provided herein may comprise a chemically reactive group, for example when said random coil polypeptide/polymer is part of a "fusion protein"/"fusion construct".
  • biosynthetic random coil polypeptide (or segment thereof) can be prepared by recombinant expression in a transformed cell in several ways according to methods well known to the person skilled in the art, for example: (i) direct expression in the cytoplasm with the help of an N-terminal Met residue / start codon; (ii) secretion via an N-terminal signal peptide, for example OmpA, PhoA ( Monteilhet (1993) Gene.
  • fusion partner is the SUMO protein, which can be cleaved by SUMO protease, as described in Examples 20 and 21.
  • Further fusion partners include, without limitation, glutathion-S-transferase, thioredoxin, a cellulose-binding domain, an albumin-binding domain, a fluorescent protein (such as GFP), protein A, protein G, an intein and the like ( Malhotra (2009) Methods Enzymol. 463:239-258 ).
  • the random coil polypeptides (or polypeptide segments) / polymers described consist predominantly of alanine and proline residues, whereas serine, threonine or asparagine, which are required for O- or N-glycosylation, are preferably absent.
  • the production of the polypeptide itself or of a biologically active protein comprising the random coil polypeptide (or polypeptide segment thereof) or, generally, a fusion protein comprising the random coil polypeptide (or polypeptide segment thereof) can result in a monodisperse product preferably devoid of post-translational modifications within the Pro-Ala sequence
  • yeast has been used for the production of approved therapeutic proteins such as insulin, granulocyte-macrophage colony stimulating factor, platelet-derived growth factor or hirudin ( Gerngross (2004) Nat.
  • CHO cells have served for the production of therapeutic proteins such as coagulation factor IX, interferone ⁇ -1a, tenecteplase ( Chu (2001) Curr. Opin. Biotechnol. 12:180-187 ) or gonadotropins, where the glycocomponent may positively influence several aspects like functional activity, folding, dimerization, secretion as well as receptor interaction, signal transduction, and metabolic clearance ( Walsh (2006) Nat. Biotechnol. 24:-1241-1252 ). Accordingly, the preparation of the inventive constructs, random coil polypeptides and conjugates in eukaryotic expression systems is also disclosed in context of the present invention.
  • therapeutic proteins such as coagulation factor IX, interferone ⁇ -1a, tenecteplase ( Chu (2001) Curr. Opin. Biotechnol. 12:180-187 ) or gonadotropins, where the glycocomponent may positively influence several aspects like functional activity, folding, dimerization, secretion as well as receptor interaction, signal transduction, and
  • conjugates and molecules comprising (i) a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues and (ii) a further molecule of interest, like a useful protein, a protein segment or a small molecule.
  • the present invention also provides for methods for the preparation or manufacure of such conjugates as well as of biosynthetic random coil polypeptides and/or molecules or conjugates comprising the same.
  • the present invention also provides for a method for the preparation and/or manufacture of a random coil polypeptide or a random coil polypeptide segment as comprised in the drug conjugates. Also methods for the preparation and/or manufacture of the biologically active protein or conjugate comprising the random coil polypeptide or the random coil polypeptide segment are provided. Furthermore, methods for the preparation and/or manufacture and/or for the preparation and/or manufacture of a polypeptide that comprises or that is an amino acid sequence that has or that mediates a biological activity and that additionally comprises said random coil polypeptide or random coil polypeptide segment are provided.
  • These methods in particular comprise (as one step) the cultuvation of the (host) cell as provided herein above and (as a further step) the isolation of said random coil polypeptide or biologically active protein and/or said biologically active protein and/or said polypeptide conjugate from the culture or from said cell.
  • This isolated random coil, a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues as well as the isolated conjugate may than be further proceesed.
  • said biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues may be chemically linked or coupled to a molecule of interest, as also shown in the appended examples.
  • the molecule of interest may be enzymatically conjugated e.g. via transglutaminase ( Besheer (2009) J Pharm Sci. 98:4420-8 ) or other enzymes ( Subul (2009) Org. Biomol. Chem. 7:3361-3371 ) to the said biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting of proline and alanine amino acid residues.
  • the random coil polypeptide (or segment thereof) and/or a protein conjugate comprising random coil polypeptide (or segment thereof) and a protein of interest, like a biologically or therapeutically active protein or a protein to be used in, e.g. diagnostic methods, can be isolated (inter alia) from the growth medium, cellular lysates, periplasm or cellular membrane fractions.
  • the present invention is not limited to (protein) conjugates that are useful in a medical or pharmaceutical setting.
  • the means and methods provided herein are also of use in other industrial areas, like, but not limited to food and beverage industry, nutrient industry, paper industry, bioreagent industry, research tool and reagent industry, industries where enzymes are to be used, cosmetic industry, oil processing and oil recovery, and the like).
  • the isolation and purification of the expressed polypeptides of the invention may be performed by any conventional means ( Scopes (1982) "Protein Purification", Springer, New York, NY ), including ammonium sulphate precipitation, affinity purification, column chromatography, gel electrophoresis and the like and may involve the use of monoclonal or polyclonal antibodies directed, e.g., against a tag fused with the biologically active protein of the invention.
  • the protein can be purified via the Strep-tag II using streptavidin affinity chromatography ( Skerra (2000) Methods Enzymol. 326:271-304 ) as described in the appended examples.
  • polypeptides of at least about 90 to 95 % homogeneity are preferred, and 98 to 99 % or more homogeneity are most preferred, in particular for phannaceutical use/applications.
  • the polypeptides of the present invention may be glycosylated or may be non-glycosylated.
  • the invention further relates to the use of the biologically active protein, the random coil polypeptide (or segment thereof) or the conjugates, like the drug conjugates of the invention, the nucleic acid molecule of the invention, the vector of the invention or the (host) cell of the invention for the preparation of a medicament, wherein said biologically active protein or drug (or any other small molecule or protein of interest) has an increased in vivo and/or in vitro stability as compared to a control molecule that does not comprise or that is not linked to a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues.
  • the present disclosure relates to a method for the treatment of diseases and/or disorders that benefit from the improved stability of said biologically active protein or drug, comprising administering the biologically active protein or drug conjugate as described herein to a mammal in need of such treatment.
  • the skilled person is readily capable of determining which disease/disorder is to be treated with a specific biologically active protein or drug conjugate of the invention.
  • Biologically active protein or a biologically active component/fragment thereof or drug Disorder/disease to be treated granulocyte colony stimulating factor cancer and/or chemotherapy related neutropenia human growth hormone growth hormone deficiency related hypoglycaemia and/or growth failure interferon ⁇ cancer, viral infection, hepatitis C interferon ⁇ auto-immune disease, multiple sclerosis interferon ⁇ viral infection tumor necrosis factor cancer interleukin-20 psoriasis ⁇ -galactosidase A Fabry disease myostatin antagonist sarcopenia gastric inhibitory polypeptide type 2 diabetes alpha-1 antitrypsin enzyme replacement therapy, cystic fibrosis, chronic obstructive pulmonary diseases, acute respiratory syndrome, severe asthma.
  • IL-1ra myocardial
  • the biologically active protein, the random coil polypeptide (or segment thereof), the drug conjugate, the nucleic acid, the vector or the cell may be used for the preparation of a medicament which preferably has or confers an increased in vivo and/or in vitro stability, in particular for the biologically active protein and/or drug component, for the treatment of hormone deficiencies or related disorders, auto-immune disease, cancer, anaemia, neovascular diseases, infectious/inflammatory diseases, thrombosis, myocardial infarction, diabetes, infertility, Gaucher's disease, hepatitis, hypoglycaemia, acromegaly, adenosine deaminase deficiency, thrombocytopenia, haemophilia, anemia, obesity, Alzheimer's disease, lipodistrophy, psoriasis, metastatic melanoma, osteoarthritis, dyslipidemia, rheumatoid arthritis, systemic lupus
  • the biologically active protein, the drug conjugate the nucleic acid, the vector or the cell is for the use as a medicament which has an increased in vivo and/or in vitro stability of said biologically active protein / drug conjugate.
  • the biologically active protein, the random coil polypeptide (or segment thereof), the drug conjugate, the nucleic acid, the vector or the cell are for use in the treatment of for the treatment of hormone deficiencies or related disorders, auto-immune disease, proliferative disorders, like cancer, anaemia, neovascular diseases, infectious and/or inflammatory diseases, thrombosis, myocardial infarction stroke, diabetes, infertility, penile dysfunction, Gaucher's disease, Fabry disease, sarcopenia, cystic fibrosis, obstructive pulmonary diseases, acute respiratory syndrome, hepatitis, hypoglycaemia, acromegaly, adenosine deaminase deficiency, thrombocytopenia,
  • the present invention also relates to the use of the nucleic acid molecules, vectors as well as transfected cells as provided herein and comprising the nucleic acid molecules or vectors of the present invention in medical approaches, like, e.g. cell based gene therapy approaches or nucleic acid based gene therapy approaches.
  • the random coil polypeptide (or polypeptide segment thereof) as provided herein, the drug conjugate that comprises the biosynthetic random coil polypeptide (or polypeptide segment thereof) and/or the nucleic acid molecule or the vector or the host cell of the present invention) is part of a composition.
  • Said composition may comprise one or more of the drug conjugates or nucleic acid molecules, vectors and/or host cells encoding and/or expressing the same.
  • Said composition may be a pharmaceutical composition, optionally further comprising a pharmaceutically acceptable carrier and/or diluent.
  • the present invention relates to the use of the herein described biologically active protein, the random coil polypeptide (or segment thereof) or the drug conjugate for the preparation of a pharmaceutical composition for the prevention, treatment or amelioration of diseases which require the uptake of such a pharmaceutical composition.
  • heterologous proteins/protein constructs comprising the inventive random coil polypeptide or polypeptide segment thereof
  • said random coil polypeptide or polypeptide segment may be per se employed in such a medical context, for example as "plasma expander” or as blood surrogate, in the amelioration, prevention and/or treatment of a disorder related to an impaired blood plasma amount or blood plasma content or in the amelioration, prevention and/or treatment of a disorder related to an impaired blood volume.
  • disorders that are treated with plasma expanders are, but not limited to, disorders affiliated with blood loss, like injuries, surgeries, burns, trauma, or abdominal emergencies, infections, dehydratations etc. Yet, such a medical use is not limited to the random coil polypeptide or polypeptide segment of this invention but can also be extended to certain drug conjugates as disclosed herein or even to certain biologically active, heterologous proteins/protein constructs.
  • the composition as described herein may be a diagnostic composition, for example an imaging reagent, optionally further comprising suitable means for detection, wherein said diagnostic composition has an increased in vivo and/or in vitro stability.
  • compositions of the invention may be in solid or liquid form and may be, inter alia, in a form of (a) powder(s), (a) tablet(s), (a) solution(s) or (an) aerosol(s). Furthermore, it is envisaged that the medicament of the invention might comprise further biologically active agents, depending on the intended use of the pharmaceutical composition.
  • compositions may be effected by different ways, e.g., by parenteral, subcutaneous, intravenous, intraarterial, intraperitoneal, topical, intrabronchial, intrapulmonary and intranasal administration and, if desired for local treatment, intralesional administration.
  • Parenteral administrations include intraperitoneal, intramuscular, intradermal, subcutaneous, intravenous or intraarterial administration.
  • the compositions of the invention may also be administered directly to the target site, e.g., by biolistic delivery to an external or internal target site, like a specifically effected organ.
  • Suitable pharmaceutical carriers, excipients and/or diluents include phosphate buffered saline solutions or other buffer solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions etc.
  • compositions comprising such carriers can be formulated by well known conventional methods.
  • Suitable carriers may comprise any material which, when combined with the biologically active protein / drug conjugate of the invention, retains its biological and/or pharmaceutical activity (see Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed, Mack Publishing Company, Easton, PA ).
  • Preparations for parenteral administration may include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • the buffers, solvents and/or excipients as employed in context of the pharmaceutical composition are preferably "physiological" as defined herein above.
  • non-aqueous solvents examples include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles may include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles may include fluid and nutrient replenishes, electrolyte replenishers (such as those based on Ringer's dextrose), and the like.
  • a pharmaceutical composition of the present invention might comprise proteinaceous carriers, like, e.g., serum albumin or immunoglobulin, preferably of human origin.
  • compositions can be administered to the subject at a suitable dose.
  • the dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depend upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Pharmaceutically active matter may be present in amounts between 1 ⁇ g and 20 mg/kg body weight per dose, e.g. between 0.1 mg to 10 mg/kg body weight, e.g. between 0.5 mg to 5 mg/kg body weight. If the regimen is a continuous infusion, it should also be in the range of 1 ⁇ g to 10 mg per kilogram of body weight per minute. Yet, doses below or above the indicated exemplary ranges also are envisioned, especially considering the aforementioned factors.
  • the pharmaceutical composition of the invention might comprise further biologically or pharmaceutically active agents, depending on the intended use of the pharmaceutical composition.
  • further biologically or pharmaceutically active agents may be e.g. antibodies, antibody fragments, hormones, growth factors, enzymes, binding molecules, cytokines, chemokines, nucleic acid molecules and drugs.
  • compositions to be used in research or as diagnostic(s) are envisaged.
  • biologically active proteins or drug conjugates comprising a random coil domain or component as defined herein, are used in a diagnostic setting.
  • the inventive biologically active protein or drug conjugate of this invention may be labelled in order to allow detection.
  • Such labels comprise, but are not limited to, radioactive labels (like [ 3 H]hydrogen [ 125 I]iodide or [ 123 I]iodide), fluorescent labels (including fluorescent proteins, like green fluorescent protein (GFP) or fluorophores, like fluorescein isothiocyanate (FITC)) or NMR labels (like gadolinium chelates).
  • radioactive labels like [ 3 H]hydrogen [ 125 I]iodide or [ 123 I]iodide
  • fluorescent labels including fluorescent proteins, like green fluorescent protein (GFP) or fluorophores, like fluorescein isothiocyanate (FITC)
  • NMR labels like gadolinium chelates
  • conjugates comprising (i) a biosynthetic random coil polypeptide or polypeptide segment comprising an amino acid sequence consisting solely of proline and alanine amino acid residues, wherein said amino acid sequence consists of at least 50 proline (Pro) and alanine (Ala) amino acid residues, and (ii) fluorescein or digoxigenin; see appended Figure 13 and 14 and the corresponding figure legend as well as illustrative Example 22.
  • the present disclosure also provides for uses of the biosynthetic random coil polypeptide as provided herein in such industrial areas.
  • Also part of this disclosure is, accordingly, a method for the production of a cosmetic, of a compound to be used in cosmetic treatments, of a food or of a beverage, said method comprising the culture of the cell comprising a nucleic acid molecule (or a vector) encoding a random coil polypeptide as defined herein or encoding a biologically active protein and/or a biologically active protein and/or a polypeptide that comprises or that is an amino acid sequence that has or that mediates an activity.
  • Such a method also includes the isolation of said random coil polypeptide, said biologically active protein and/or said biologically active protein or said polypeptide that comprises or that is an amino acid sequence that has or that mediates an activity, like a biological activity, and that additionally comprises said random coil polypeptide or random coil polypeptide segment from the culture or from said cell.
  • conjugates of interest can be produced, for example conjugates which are useful in different areas of industry, like in the oil or paper industry.
  • kits comprising the drug conjugate, the nucleic acid molecule encoding said biologically active protein and/or encoding said polypeptide that comprises or that is an amino acid sequence that has or that mediates an activity (for example a biological activity), the vector comprising said nucleic acid molecule or the cell comprising said nucleic acid or said vector as described herein.
  • the kit of the present invention may further comprise, (a) buffer(s), storage solutions and/or additional reagents or materials required for the conduct of medical or scientific assays and purposes.
  • parts of the kit can be packaged individually in vials or bottles or in combination in containers or multicontainer units.
  • the kit may be advantageously used, inter alia, for carrying out the method of the invention and could be employed in a variety of applications referred herein, e.g., as research tools or as medical tools. Additionally, the kit may contain means for detection suitable for scientific or medical purposes.
  • the manufacture of the kits preferably follows standard procedures which are known to the person skilled in the art.
  • Example 1 Gene synthesis for Pro/Ala amino acid polymers/polypeptides.
  • Pro/Ala amino acid repeats consisting of Pro and Ala residues are depicted herein as Pro/Ala or "PA”.
  • Gene fragments encoding a repetitive polymer sequence comprising Pro and Ala (PA#1 which corresponds to SEQ ID NO: 1) were obtained by hybridisation of the two complementary oligodeoxynucleotides (SEQ ID NO: 17 and SEQ ID NO: 18) shown in Fig. 1 , followed by concatamer formation in a directed manner via DNA ligation of their mutually compatible but non-palindromic sticky ends.
  • Oligodeoxynucleotides were purchased from ThermoScientific (Ulm, Germany) and purified by preparative urea polyacrylamide gel electrophoresis.
  • Fig. 1 The nucleic acid sequences of the oligodesoxynucleotides are depicted in Fig. 1 (SEQ ID NOs 17 and 18 comprising an additional GCC codon for alanine, which becomes part of the following PA#1 sequence repeat upon ligation of the corresponding sticky ends.
  • Enzymatic phosphorylation was performed by mixing 200 pmol of both oligodeoxynucleotides in 100 ⁇ l 50 mM Tris/HCl pH 7.6, 10 mM MgCl 2 , 5 mM DTT, ImM ATP and incubation for 30 min at 37 °C in the presence of 10 u polynucleotide kinase (MBI Fermentas, St. Leon-Rot, Germany).
  • Example 2 Construction of pFab-PA#1(200) as expression vector for a Fab-PA#1 fusion protein.
  • Example 3 Construction of pASK-PA#1(200)-IFNa2b as an expression vector for a PA#1(200)-IFNa2b fusion protein.
  • plasmid was prepared and the presence of the correct insert was confirmed by restriction analysis.
  • the resulting plasmid was designated pPA#1(200)-IFNa2b (SEQ ID NO: 37) ( Fig. 2F ).
  • Example 4 Bacterial production and purification of fusion proteins between an Fab fragment and a genetically encoded PA#1 polymer/polypeptide.
  • Example 5 Bacterial production and purification of fusion proteins between IFNa2b and a genetically encoded PA#1 polymer/polypeptide.
  • Periplasmic extraction in the presence of 500 mM sucrose, 1 mM EDTA, 100 mM Tris/HCl pH 8.0 containing 50 ⁇ g/ml lysozyme was performed as described elsewhere (Breustedt (2005) loc. cit.) and followed by purification via the Strep-tag II using streptavidin affinity chromatography ( Schmidt (2007) Nat. Protoc. 2:1528-1535 ) in the presence of 150 mM NaCl, 1 mM EDTA, 100 mM Tris/HCl, pH 8.0.
  • Example 6 Measurement of the hydrodynamic volume for the recombinant fusion protein between a Fab fragment and a genetically encoded PA#1 polymer of 200 residues by analytical gel filtration.
  • Size exclusion chromatography was carried out on a Superdex S200 HR 10/300 GL column (GE Healthcare Europe, Freiburg, Germany) at a flow rate of 1 ml/min using an ⁇ kta Purifier 10 system (GE Healthcare) with PBS (115 mM NaCl, 4 mM KH 2 PO 4 , 16 mM Na 2 HPO 4 ; pH 7.4) as running buffer. 250 ⁇ l samples of the purified Fab fragment and its 200 residue PA#1 fusion, obtained from the metal affinity affinity chromatography as described in Example 4, were individually applied at a concentration of 0.25 mg/ml in PBS. Both proteins eluted in a single homogenous peak as shown in Fig. 4A .
  • globular proteins (Sigma, Deisenhofen, Germany) were applied in PBS at protein concentrations between 0.2 mg/ml and 0.5 mg/ml: cytochrome c, 12.4 kDa; carbonic anhydrase, 29.0 kDa; ovalbumin, 43.0 kDa; bovine serum albumin, 66.3 kDa; alcohol dehydrogenase, 150 kDa; ⁇ -amylase, 200 kDa; apo-ferritin, 440 kDa.
  • cytochrome c 12.4 kDa
  • carbonic anhydrase 29.0 kDa
  • ovalbumin 43.0 kDa
  • bovine serum albumin 66.3 kDa
  • alcohol dehydrogenase 150 kDa
  • ⁇ -amylase 200 kDa
  • apo-ferritin 440 kDa.
  • the fusion protein with the 200 residue PA#1 polymer/polypeptide exhibited a significantly larger size than corresponding globular proteins with the same molecular weight.
  • the apparent size increase for Fab-PA#1(200) was 7.4-fold compared with the unfused Fab fragment whereas the true mass was only larger by 1.3-fold. This observation clearly indicates a much increased hydrodynamic volume conferred to the biologically active Fab fragment by the Pro/Ala polypeptide segment according to this invention.
  • Example 7 Measurement of the hydrodynamic volume for the recombinant fusion protein between IFNa2b and a genetically encoded PA#1 polymer of 200 residues by analytical gel filtration.
  • Size exclusion chromatography was carried out with IFNa2b and PA#1(200)-IFNa2b on a Superdex S200 HR 10/300 GL column (GE Healthcare) at a flow rate of 1 ml/min using an ⁇ kta Purifier 10 system (GE Healthcare) similarly as described in Example 6. Both proteins eluted in a single homogenous peak as shown in Fig. 4C .
  • the fusion protein with the 200 residue PA#1 polymer/polypeptide exhibited a significantly larger size than corresponding globular proteins with the same molecular weight ( Fig. 4D ).
  • the apparent size increase for PA#1(200)-IFNa2b was 10.2-fold compared with the unfused IFNa2b protein whereas the true mass was only larger by 1.8-fold. This observation clearly indicates a much increased hydrodynamic volume conferred to the biologically active interferon by the Pro/-Ala polymer/polypeptide according to this invention.
  • Example 8 Detection of random coil conformation for the biosynthetic PA#1 polymer fused to a Fab fragment via circular dichroism spectroscopy.
  • ⁇ M ⁇ obs c ⁇ d
  • ⁇ obs denotes the measured ellipticity
  • c protein concentration [mol/l]
  • d path length of the quartz cuvette [cm].
  • Kaleidagraph Synergy Software, Reading, PA
  • the measured circular dichroism (CD) spectrum for the recombinant Fab was in accordance with the ⁇ -sheet dominated immunglobuline fold, whereas the spectrum for the Fab-PA#1(200) fusion protein revealed a significant contribution of random coil conformation ( Fig. 5A ).
  • the molar difference CD spectrum with respect to the unfused Fab fragment was calculated ( Fig. 5B ) by subtraction of the latter spectrum from the one for Fab-PA#1(200).
  • a strong minimum around 200 nm which is characteristic of random coil conformation, was observed.
  • the Pro/Ala sequence as part of the recombinant fusion protein appears to be present as a random coil polymer under physiological buffer conditions.
  • Example 9 Detection of random coil conformation for the genetically encoded PA#1 polymer fused to IFNa2b via circular dichroism spectroscopy.
  • Example 10 Quantitative analysis of the secondary structure of the Fab fragment, of IFNa2b and of their 200 residue PA#1 polymer fusions.
  • the secondary structure content of the Fab fragment, Fab-PA# 1(200), IFNa2b, and PA#1(200)-IFNa2b was individually quantified from the corresponding CD spectra measured in Examples 8 and 9 using the secondary structure deconvolution program CDNN ver. 2.1 ( Böhm (1992) Protein Eng.
  • the fraction of unstructured conformation for the whole protein clearly increases if the PA#1 polymer is fused to IFNa2b.
  • the difference CD spectrum for the Pro/Ala polypeptide segment reveals a clear random coil conformation.
  • Analysis of the secondary structure shows the presence of a high fraction of unstructured conformations (comprising random coil and ⁇ -turn) which nearly comprise 100 % of the total secondary structure.
  • Example 11 Construction of pASK75-His6-PA#1(200)-hGH as an expression vector for a His6-PA#1(200)-hGH fusion protein.
  • plasmid was prepared and the presence of the correct insert was confirmed by restriction analysis.
  • the resulting plasmid was designated pASK75-His6-PA#1(200)-hGH (SEQ ID NO: 46) ( Fig. 6C ).
  • Example 12 Construction of an expression vector for the secretory production of human growth hormone fused with a 200 residue PA#1 polymer/polypeptide in Chinese hamster ovary cells.
  • the vector pASK75-His6-PA#1(200)-hGH (SEQ ID NO: 46), a derivative of pASK75 (Skerra (1994) loc. cit.), allowing prokaryotic production of the hGH PA#1 fusion protein, was cut with Nhe I and Hind III. This fragment was purified via agarose gel electrophoresis and ligated with the correspondingly cut vector pCHO (SEQ ID NO: 50). After transformation of E. coli XL1-Blue (Bullock (1987) loc. cit.), plasmid was prepared and the correct insertion of the fragment was verified via restriction analysis.
  • the resulting plasmid which codes for the hGH signal peptide fused to the His 6 tag, a PA#1(200) polypeptide segment, and the human growth hormone (hGH), was designated pCHO-PA#1(200)-hGH SEQ ID NO: 48) and is depicted in Fig. 6D .
  • Example 13 Secretory production of a fusion protein between human growth hormone (hGH) and the genetically encoded PA#1 polymer in CHO cells.
  • hGH human growth hormone
  • CHO-K1 cells ATCC No. CCL-61 were cultured in Quantum 263 medium (PAA Laboratories, Cölbe, Germany) in a 100 mm plastic dish until 50 % confluency was reached.
  • Cells were transfected with 8 ⁇ g pCHO-PA#1(200)-hGH (SEQ ID NO: 48) or, for control, pCHO-hGH (SEQ ID NO: 49), a similar plasmid encoding hGH without the PA#1(200) sequence, using the Nanofectin Kit (PAA Laboratories, Cölbe, Germany).
  • cell culture medium was exchanged by 7 ml Opti-MEM®-I reduced serum medium (Invitrogen, Darmstadt, Germany) and cells were incubated at 37 °C in a humidified atmosphere with 5 % CO2. After two days, 20 ⁇ l of the cell culture supernatant was taken and diluted with 5 ⁇ l SDS-PAGE loading buffer containing ⁇ -mercaptoethanol. After 5 min heating at 95 °C, 15 ⁇ l of each sample was subjected to 12 % SDS-PAGE.
  • a nitrocellulose membrane (Schleicher & Schuell, Dassel, Germany) by means of a semi-dry blotting apparatus, the membrane was washed 3 times for 15 min with 10 ml PBST (PBS containing 0.1 % v/v Tween 20). The membrane was incubated with 10 ml of a 1:1000 dilution of anti human growth hormone antibody ab1956 conjugated with horse radish peroxidase (Abcam, Cambridge, UK).
  • Example 14 Bacterial production and purification of fusion proteins between hGH and a genetically encoded PA#1 polymer/polypeptide.
  • hGH Human growth hormone
  • PA#1(200)-hGH (calculated mass: 39.6 kDa)
  • PA#1(400)-hGH (calculated mass: 55.8 kDa)
  • PA#1(600)-hGH (calculated mass: 72.0 kDa) were produced in E. coli KS272 harboring the corresponding expression plasmids from Example 11 or their derivatives with a double (encoding 400 residues) or triple (600 residues) PA#1 sequence cassette, respectively.
  • Example 15 Measurement of binding affinity of human growth hormone and its PA#1 polymer fusions towards the extracellular domain of human growth hormone receptor using surface plasmon resonance.
  • hGH and its PA#1 polypeptide fusions to a human growth hormone receptor Fc fusion protein was determined via surface plasmon resonance (SPR) real time measurements on a Biacore 2000 system (GE Healthcare).
  • SPR surface plasmon resonance
  • 15 ⁇ l mouse anti-human IgG-Fc capture antibody Jackson Immuno Research
  • 10 mM Na-acetate pH 5.0 was immobilized to the surface of two flow channels of a CMDP chip (XanTec bioanalytics) using an amine coupling kit (GE Healthcare). This resulted in ca. 2700 response units (RU).
  • the sensograms were corrected by double subtraction of the corresponding signals measured for the channel without immobilized receptor and an averaged baseline determined from several buffer blank injections ( Myszka (1999) Mol. Recognit. 12: 279-284 ).
  • Kinetic data evaluation was performed by a global fit of the traces from at least seven different sample injections according to the 1:1 Langmuir binding model using BIAevaluation software version 3.1 (GE Healthcare).
  • Example 16 Detection of prolonged plasma half-life in vivo for the recombinant fusion proteins between a Fab fragment and genetically encoded PA#1 polymers.
  • mice Female BALB/c mice (SPF stock breeding; TU München, Freising, Germany) were intravenously injected according to the following Table: Group A B D Test item Fab Fab-PA#1(200) Fab-PA#1(600) Administration route intravenous Dose [mg/kg b.w.] 5.0 5.0 5.0 Concentration [mg/ml] 1.0 1.0 1.0 Application volume [ml/kg b.w.] 5.0 No. of animals/group 9 9 9 No. of blood sampling time points 12 12 12 No. of animals/sampling time point 3 3 3 3 No. of blood samplings/animal 4/1 4/1 4/1
  • the total volume of intravenously administered test item was calculated according to the individual body weight (b.w.) recorded on the day of administration (e.g. an animal with 20 g body weight received 100 ⁇ l of 1 mg/ml test item). Blood sampling was performed according to the following Table: Time points for blood sampling after injection Test item Subgroup 10 min 30 min 1 h 2 h 3 h 4 h 6 h 8 h 12 h 24 h 36 h 48 h Fab Fab-PA# 1 (200) Fab-PA# 1 (600) 1 X X X X X X X X X X X 2 X X X X X X X X X X X X X X X X X X X 3 X X X X X X X X X X X X X X X X X X X X X X X X X X X X X
  • Plasma samples (approximately 50 ⁇ l) were taken from the tail vene and stored at 4 °C for 30 min. After centrifugation for 10 min at 10 000 g and 4 °C the supernatant (plasma) was immediately frozen and stored at -20 °C.
  • the wells of a 96 well microtiter plate were coated overnight at 4 °C with 50 ⁇ l of a 10 ⁇ g/ml solution of recombinant Her2/ErbB2 ectodomain antigen in 50 mM NaHCO 3 pH 9.6. Then, the wells were blocked with 200 ⁇ l of 3 % (w/v) BSA in PBS for 1 h and washed three times with PBS/T (PBS containing 0.1 % (v/v) Tween 20).
  • the plasma samples were applied in dilution series in PBS/T containing 0.5 % (v/v) mouse plasma from an untreated animal and incubated for 1 h. The wells were then washed three times with PBS/T and incubated for 1 h with 50 ⁇ l of a 1:1000 diluted solution of an anti-human C ⁇ antibody alkaline phosphatase conjugate in PBS/T.
  • c 0 is the total blood concentration at time point zero while c ⁇ is the concentration amplitude for the distribution phase.
  • Figure 8 depicts the pharmacokinetics for the three test items in BALB/c mice. While the recombinant Fab shows a rapid blood clearance with an elimination half-life of just ca. 1.3 h, the Fab-PA#1(200) and Fab-PA#1(600) fusion proteins have a more than 3-fold and 29-fold extended half-life with corresponding values of ca. 4.1 h and 38.8 h, respectively. These data prove that the in vivo plasma half-life of a Fab fragment is significantly prolonged due to fusion with a Pro/Ala polymer/polypeptide, whereby the half-life becomes longer with increasing length of the amino acid polymer.
  • Example 17 Gene synthesis for P1A1 and P1A3 amino acid polymers/polypeptides and construction of pFab-P1A1(200) and pFab-P1A3(200) as expression vectors for Fab-P1A1(200) and Fab-P1A3(200) fusion proteins.
  • Gene fragments encoding a repetitive polymer sequence comprising the Pro/Ala polypeptides/polymers P1A1 (SEQ ID NO: 51) and P1A3, also designated PA#3, (SEQ ID NO: 3) were obtained by hybridisation of pairs of complementary oligodeoxynucleotides, respectively, SEQ ID NO: 52 and SEQ ID NO: 53 for P1A1 and SEQ ID NO: 54 and SEQ ID NO: 55 for P1A3 as described in Example 1.
  • pFab-P1A1(200) (Seq ID NO: 58) and pFab-P1A3(200) (Seq ID NO: 59) coding for Fab fragments with the corresponding Pro/Ala polymers/polypeptide segments of 200 residues at the C-terminus of the light chain (LC) (amino acid sequence of LC Fab-P1A1(200): SEQ ID NO: 56; amino acid sequence of LC Fab-P1A3(200): SEQ ID NO: 57) were constructed in an analogous manner to pFab-PA# 1(200), which has been described in Example 2.
  • LC light chain
  • Example 18 Measurement of the hydrodynamic volume for the recombinant fusion protein between a Fab fragment and a genetically encoded P1A1 or P1A3 polypeptide/polymer by analytical gel filtration.
  • the fusion proteins with the 200 residue P1A1 or P1A3 polymers/polypeptides exhibited significantly larger sizes than the corresponding unfused Fab fragment.
  • the apparent size increase for Fab-P1A1(200) and Fab-P1A3(200) was 5.8-fold and 5.2-fold, respectively, compared with the Fab fragment (cf. Fig. 4B ) whereas the true mass was only larger by 1.4-fold and 1.3-fold. This observation clearly indicates a much increased hydrodynamic volume conferred to the biologically active Fab fragment by the biosynthetic P1A1 and P1A3 polypeptide segments according to this invention.
  • Example 19 Detection of random coil conformation for the biosynthetic P1A1 and P1A3 polymers/polypeptides fused to a Fab fragment via circular dichroism (CD) spectroscopy.
  • CD circular dichroism
  • CD spectra for Fab-P1A1(200) and Fab-P1A3(200) were recorded as described in Example 8 using 4.2 and 6.5 ⁇ M protein solutions, respectively, prepared similary as described in Example 4 using 50 mM K 2 SO 4 , 20 mM K-phosphate pH 7.5 as buffer.
  • Example 20 Construction of pSUMO-PA#1(200) as expression vector for a His(6)-SUMO-PA#1(200) fusion protein.
  • the 5'-primer introduced an Nde I restriction site, containing a Met start codon (ATG) and an additional Lys codon, as well as the His6-tag encoding sequence while the 3'-primer introduced a Hind III and Sap I restriction site into the PCR product.
  • the resulting DNA fragment was digested with Nde I and Hind III and ligated with a correspondingly digested derivative of the plasmid pSA1 ( Schmidt (1994) J. Chromatogr. 676: 337-345 ), wherein the Sap I restriction site had been eliminated by silent mutation.
  • the resulting plasmid was cut with Sap I , dephosphorylated with shrimp alkaline phosphatase, and ligated with the gene fragment encoding the 200 residue PA#1 polypeptide segment excised from the plasmid pFab-PA#1(200) (described in Example 2) by restriction digest with Sap I (in an analogous way as exemplified in Figure 2E ).
  • the resulting plasmid was designated pSUMO-PA#1(200) (SEQ ID NO: 60) and is depicted in Figure 12A .
  • Example 21 Bacterial expression and isolation of a genetically encoded PA#1(200) polymer/polypeptide.
  • the PA#1(200) polypeptide (calculated mass: 16.1 kDa) was initially produced as fusion protein with the small ubiquitin-like modifier (SUMO) protein (calculated mass: 12.2 kDa) in the cytoplasm of E. coli BLR(DE3) (NEB, Ipswich, MA, USA) harboring the expression plasmid pSUMO-PA#1(200) (described in Example 21) together with the plasmid pLysE ( Studier (1991) J. Mol. Biol. 219: 37-44 ), which suppresses the the T7 promoter.
  • SUMO small ubiquitin-like modifier
  • Bacterial production was performed at 30 °C in shake flask cultures with 2 L LB medium containing 2.5 g/L D-glucose, 0.5 g/L L-proline, 100 mg/l ampicillin, and 30 mg/l chloramphenicol. Recombinant gene expression was induced by addition of isopropyl- ⁇ -D-thiogalactopyranoside (IPTG) to a final concentration of 0.5 mM. Bacteria were harvested 3 h after induction, resuspended in 100 mM NaCl, 40 mM Na-phosphate pH 7.5 and lysed using a French pressure cell (Thermo Scientific, Waltham, MA, USA). After centrifugation (15 min, 15000 g) of the lysate no inclusion bodies were observed.
  • IPTG isopropyl- ⁇ -D-thiogalactopyranoside
  • the supernatant containing the soluble fusion protein was incubated at 70 °C for 15 min and centrifuged (15 min, 15000 g) to remove thermally unstable host cell proteins.
  • the His(6)-SUMO-PA#1(200) fusion protein was purified from the supernatant via IMAC ( Skerra (1994) Gene 141: 79-84 ) using a 12 ml Ni 2 + charged HisTrap high performance column (GE Healthcare) connected to an ⁇ kta purifier system (GE Healthcare) and eluted with an imidazole gradient from 0 to 150 mM in 500 mM NaCl, 40 mM Na-phosphate pH 7.5.
  • the biosynthetic PA#1(200) polypeptide was liberated from the fusion protein by site specific proteolytic cleavage (downstream of a Gly-Gly motif preceding the Pro/Ala polypeptide segment) with 2 U/mg Ubl-specific protease 1 from Saccharomyces cerevisiae (Invitrogen, Carlsbad, CA, USA) for 1 h at 30 °C in cleavage buffer (0.2 w/v % Igepal, 1 mM DTT, 150 mM NaCl, 50 mM Tris-HCl pH 8.0).
  • the cleavage process was checked by SDS-PAGE ( Figure 12B ) using a high molarity Tris buffer system ( Fling (1986) Anal. Biochem. 155: 83-88 ).
  • a high molarity Tris buffer system Fling (1986) Anal. Biochem. 155: 83-88 ).
  • the reaction mixture was subjected to another IMAC using a 5 ml Ni 2 + charged HisTrap high performance column (GE Healthcare) and 500 M NaCl, 20 mM phosphate, pH 7.5 as running buffer. This time the flow-through contained the pure biosynthetic PA#1(200) polypeptide ( Figure 13 E) .
  • biosynthetic PA#1(200) polypeptide/polymer (SEQ ID NO: 61) prepared in this manner comprises altogether 201 amino acid residues, which arise from the encoded combined gene product of 10 ligated double-stranded oligodeoxynucleotide building blocks, each encoding 20 amino acid residues, as shown in Figure 1 , and an additional Ala residue encoded by the triplet DNA overhang of the downstream Sap I restriction site that was used for cloning.
  • the unpurified proteolytic cleavage reaction mixture of the His(6)-SUMO-PA# 1(200) fusion protein from Example 21 was twice dialysed at 4 °C against 50 mM NaHCO 3 pH 8.3 and incubated at room temperature for 1 h after mixing with a 10-fold molar excess of a solution of 6-[fluorescein-5(6)-carboxamido] hexanoic acid N-hydroxysuccinimide ester (Fluorescein-NHS ester; Sigma-Aldrich) in dry dimethylformamide (DMF).
  • DMF dry dimethylformamide
  • the present invention relates to and refers to the following exemplified sequences, whereby the appended sequence listing is presented as part of the description and is, accordingly a part of this specification.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Diabetes (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Pulmonology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Rheumatology (AREA)
  • Obesity (AREA)
  • Toxicology (AREA)
  • Communicable Diseases (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Zoology (AREA)
  • Heart & Thoracic Surgery (AREA)
EP11725656.0A 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses Active EP2571510B1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP11725656.0A EP2571510B1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses
SI201131597T SI2571510T1 (sl) 2010-05-21 2011-05-20 Biosintetični prolin/alaninski naključno zviti polipeptidi in njihove uporabe
EP18184601.5A EP3473261A1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses
PL11725656T PL2571510T3 (pl) 2010-05-21 2011-05-20 Biosyntetyczne polipeptydy prolinowo/alaninowe o strukturze kłębka statystycznego i ich zastosowania

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP10163564 2010-05-21
US201061428016P 2010-12-29 2010-12-29
EP11725656.0A EP2571510B1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses
PCT/EP2011/058307 WO2011144756A1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP18184601.5A Division EP3473261A1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses

Publications (2)

Publication Number Publication Date
EP2571510A1 EP2571510A1 (en) 2013-03-27
EP2571510B1 true EP2571510B1 (en) 2018-08-08

Family

ID=42629445

Family Applications (2)

Application Number Title Priority Date Filing Date
EP18184601.5A Withdrawn EP3473261A1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses
EP11725656.0A Active EP2571510B1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP18184601.5A Withdrawn EP3473261A1 (en) 2010-05-21 2011-05-20 Biosynthetic proline/alanine random coil polypeptides and their uses

Country Status (19)

Country Link
US (3) US9221882B2 (pt)
EP (2) EP3473261A1 (pt)
JP (2) JP5828889B2 (pt)
KR (1) KR101872541B1 (pt)
CN (2) CN102883734B (pt)
AU (1) AU2011254564B2 (pt)
BR (1) BR112012029577B1 (pt)
CA (1) CA2794614C (pt)
DK (1) DK2571510T3 (pt)
EA (1) EA024755B1 (pt)
ES (1) ES2691642T3 (pt)
HK (1) HK1219902A1 (pt)
LT (1) LT2571510T (pt)
MX (2) MX357674B (pt)
NZ (1) NZ602522A (pt)
PL (1) PL2571510T3 (pt)
SG (1) SG185440A1 (pt)
SI (1) SI2571510T1 (pt)
WO (1) WO2011144756A1 (pt)

Families Citing this family (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX357674B (es) 2010-05-21 2018-07-18 Xl Protein Gmbh Polipeptidos biosinteticos de espira aleatoria de prolina/alanina y sus usos.
US20140323402A1 (en) * 2011-08-12 2014-10-30 Ascendis Phama A/S Protein Carrier-Linked Prodrugs
KR102057356B1 (ko) 2012-02-27 2019-12-18 아뮤닉스 파마슈티컬스, 인크. Xten 콘주게이트 조성물 및 그의 제조 방법
CA2986512C (en) 2012-04-04 2022-05-17 Halozyme, Inc. Combination therapy with an anti-hyaluronan agent and a tumor-targeted taxane
EP2948777B1 (en) * 2013-01-22 2019-06-26 Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. Dipeptide-repeat proteins as therapeutic target in neurodegenerative diseases with hexanucleotide repeat expansion
JP6259207B2 (ja) * 2013-06-17 2018-01-10 雪印メグミルク株式会社 エラスチン産生促進剤
JP6259208B2 (ja) * 2013-06-17 2018-01-10 雪印メグミルク株式会社 ヒアルロン酸産生促進剤
JP6259209B2 (ja) * 2013-06-17 2018-01-10 雪印メグミルク株式会社 コラーゲン産生促進剤
CA2929201A1 (en) 2013-11-11 2015-05-14 Ascendis Pharma Relaxin Division A/S Relaxin prodrugs
US9618507B2 (en) 2014-02-24 2017-04-11 Betanien Hospital Methods of treating rheumatoid arthritis
WO2015132004A1 (en) 2014-03-05 2015-09-11 Merz Pharma Gmbh & Co. Kgaa Novel recombinant clostridial neurotoxins with increased duration of effect
PL228341B1 (pl) * 2014-04-21 2018-03-30 Bio Ventures Inst Spolka Z Ograniczona Odpowiedzialnoscia Sposób uzyskiwania białka poliepitopowego oraz wektor DNA do realizacji tego sposobu
AU2015308987B2 (en) * 2014-08-26 2021-02-18 Betanien Hospital Methods, agents and compositions for treatment of inflammatory conditions
ES2896971T3 (es) 2014-11-21 2022-02-28 Ascendis Pharma Endocrinology Div A/S Formas de administración de hormona del crecimiento de acción prolongada
WO2016193380A1 (en) 2015-06-02 2016-12-08 Novo Nordisk A/S Insulins with polar recombinant extensions
TW201718627A (zh) 2015-06-11 2017-06-01 梅茲製藥有限兩合公司 重組梭菌神經毒素及其使用與形成方法、包括其之醫藥組合物及對應其之前驅物、編碼前驅物之核酸序列及其獲得方法與前驅物之形成方法、載體與包括核酸序列之重組宿主細胞
EP3347874B1 (en) * 2015-09-09 2021-05-12 Technische Universiteit Eindhoven Imaging of dispersion and velocity of contrast agents
MA43348A (fr) 2015-10-01 2018-08-08 Novo Nordisk As Conjugués de protéines
AU2016336150B2 (en) * 2015-10-08 2023-08-10 The Governors Of The University Of Alberta Hepatitis C virus E1/E2 heterodimers and methods of producing same
MX2018007680A (es) * 2015-12-22 2018-11-14 Xl Protein Gmbh Acidos nucleicos que codifican secuencias repetitivas de aminoacidos ricas en residuos de prolina y alanina que tienen secuencias nucleotidicas con baja repetitividad.
ES2968038T3 (es) 2015-12-23 2024-05-06 Univ Johns Hopkins Agonista GLP-1R de acción prolongada como terapia de afecciones neurológicas y neurodegenerativas
NZ743488A (en) 2016-01-08 2023-02-24 Ascendis Pharma Growth Disorders As Controlled-release cnp agonists with low initial npr-b activity
IL259658B1 (en) 2016-01-08 2024-02-01 Ascendis Pharma Growth Disorders As Controlled-release CNP agonists with reduced side effects
AU2017205268B2 (en) 2016-01-08 2022-02-17 Ascendis Pharma Growth Disorders A/S CNP prodrugs with large carrier moieties
LT3400019T (lt) 2016-01-08 2022-12-12 Ascendis Pharma Growth Disorders A/S Cnp provaistai su prie žiedo fragmento prijungtu nešikliu
KR102373744B1 (ko) 2016-01-08 2022-03-15 아센디스 파마 그로우쓰 디스오더스 에이/에스 낮은 npr-c 결합을 갖는 제어 방출성 cnp 작용제
BR112018011152A2 (pt) 2016-01-08 2018-11-21 Ascendis Pharma Growth Disorders As agonistas de cnp de liberação controlada com estabilidade em nep aumentada
US11078472B2 (en) 2016-01-20 2021-08-03 Merz Pharma Gmbh & Co., Kgaa Recombinant clostridial neurotoxins with increased duration of effect
IL301616A (en) 2016-03-01 2023-05-01 Ascendis Pharma Bone Diseases As PTH medications
JP2019507118A (ja) 2016-03-02 2019-03-14 メルツ ファルマ ゲーエムベーハー ウント コンパニー カーゲーアーアー ボツリヌス毒素を含む組成物
WO2017173346A1 (en) * 2016-04-01 2017-10-05 Arizona Board Of Regents On Behalf Of The University Of Arizona Par3 mimetic peptides and uses thereof
CN109310780A (zh) * 2016-05-04 2019-02-05 纳维格蛋白质有限公司 包含肽接头的用于化学部分位点-特异性偶联的靶向化合物
AU2017295938C1 (en) 2016-07-13 2021-10-07 Ascendis Pharma A/S Conjugation method for carrier-linked prodrugs
PL3518960T3 (pl) 2016-09-29 2023-12-27 Ascendis Pharma Bone Diseases A/S Schemat dawkowania związku pth o kontrolowanym uwalnianiu
US11564974B2 (en) 2016-09-29 2023-01-31 Ascendis Pharma Growth Disorders A/S Combination therapy with controlled-release CNP agonists
US20190224329A1 (en) 2016-09-29 2019-07-25 Ascendis Pharma Bone Diseases A/S Incremental Dose Finding in Controlled-Release PTH Compounds
DK3518961T3 (da) 2016-09-29 2023-04-24 Ascendis Pharma Bone Diseases As PTH-forbindelser med lave forhold mellem top og bund
US11807671B2 (en) * 2016-11-16 2023-11-07 Auckland Uniservices Limited Methods for protein ligation and uses thereof
CN110505885A (zh) 2017-04-05 2019-11-26 诺和诺德股份有限公司 寡聚体延伸的胰岛素-Fc缀合物
EP3642222A1 (en) 2017-06-20 2020-04-29 Merz Pharma GmbH & Co. KGaA Novel recombinant botulinum toxin with increased duration of effect
US10174302B1 (en) 2017-06-21 2019-01-08 Xl-Protein Gmbh Modified L-asparaginase
CN110891612A (zh) * 2017-06-21 2020-03-17 Xl-蛋白有限责任公司 蛋白质药物与p/a肽的缀合物
EP3642340B1 (en) * 2017-06-21 2024-03-20 Jazz Pharmaceuticals Ireland Limited Modified l-asparaginase
EP3418383A1 (en) * 2017-06-21 2018-12-26 XL-protein GmbH Modified l-asparaginase
WO2019007509A1 (en) 2017-07-06 2019-01-10 Merz Pharma Gmbh & Co. Kgaa NOVEL RECOMBINANT BOTULINUM NEUROTOXINS WITH INCREASED DURATION
EP3483619A1 (en) * 2017-11-13 2019-05-15 Technische Universität München Automated noninvasive determining the sex of an embryo of and the fertility of a bird's egg
EP3713595A1 (en) * 2017-11-22 2020-09-30 Merz Pharma GmbH & Co. KGaA Novel recombinant botulinum toxin with increased duration of effect
US20190184028A1 (en) * 2017-12-14 2019-06-20 Janssen Biotech, Inc. Targeting with firbronectin type iii like domain molecules
GB201804092D0 (en) * 2018-03-14 2018-04-25 Imperial Innovations Ltd Methods and compositions
AU2019246390A1 (en) 2018-03-28 2020-08-06 Ascendis Pharma A/S Conjugates
IL308797A (en) 2018-03-28 2024-01-01 Ascendis Pharma Oncology Div A/S IL-2 conjugates
TWI724392B (zh) * 2018-04-06 2021-04-11 美商美國禮來大藥廠 生長分化因子15促效劑化合物及其使用方法
BR112020022306A2 (pt) 2018-05-18 2021-02-23 Ascendis Pharma Bone Diseases A/S dose de partida de conjugados de pth
EP3856764A4 (en) 2018-09-27 2022-11-02 Xilio Development, Inc. MASKED CYTOKINE POLYPEPTIDES
US20220025344A1 (en) 2018-11-26 2022-01-27 Novartis Ag Lpl-gpihbp1 fusion polypeptides
EP3908832A4 (en) * 2019-01-10 2023-07-19 Roswell Biotechnologies Inc. CONDUCTIVE SYNTHETIC PEPTIDES FOR MOLECULAR ELECTRONICS
EP3923905A1 (en) 2019-02-11 2021-12-22 Ascendis Pharma Bone Diseases A/S Liquid pharmaceutical formulations of pth conjugates
JP2022520193A (ja) 2019-02-11 2022-03-29 アセンディス ファーマ グロース ディスオーダーズ エー/エス Cnpコンジュゲートの乾燥医薬製剤
CA3131817A1 (en) 2019-03-04 2020-09-10 Ascendis Pharma Endocrinology Division A/S Long-acting growth hormone dosage forms with superior efficacy to daily somatropin
EP3714909A1 (en) * 2019-03-28 2020-09-30 Industrie Biomediche Insubri S.A. Improved bone implant matrix comprising proline-rich peptide and method of preparing the same
US10784093B1 (en) * 2019-04-04 2020-09-22 Thermo Finnigan Llc Chunking algorithm for processing long scan data from a sequence of mass spectrometry ion images
CA3143436A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma A/S Conjugates of .pi.-electron-pair-donating heteroaromatic nitrogen-comprising compounds
EP3986471A1 (en) 2019-06-21 2022-04-27 Ascendis Pharma A/S Conjugates of an electron-donating nitrogen or tertiary amine comprising compounds
WO2020254606A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma A/S Conjugates of heteroaromatic nitrogen-comprising compounds
MX2022002337A (es) 2019-08-27 2022-06-08 Tonix Pharma Ltd Polipéptidos de tff2 modificados.
WO2021113434A1 (en) * 2019-12-03 2021-06-10 Rodan & Fields, Llc Peptides and compositions for inhibiting hair growth
EP4084872A1 (en) 2020-01-03 2022-11-09 Ascendis Pharma A/S Conjugates undergoing intramolecular rearrangements
US20230042670A1 (en) 2020-01-13 2023-02-09 Ascendis Pharma Bone Diseases A/S Hypoparathyroidism Treatment
GB202007106D0 (en) * 2020-05-14 2020-07-01 Ucl Business Plc Cyclosporine analogues
IL298642A (en) 2020-06-03 2023-01-01 Ascendis Pharma Oncology Div A/S il-2 sequences and uses thereof
AU2021319863A1 (en) 2020-08-05 2023-02-16 Ascendis Pharma A/S Conjugates comprising reversible linkers and uses thereof
TW202228784A (zh) 2020-09-23 2022-08-01 奧地利商艾柏力維亞生技有限公司 用以於一患者中螯合非預期的抗peg抗體的化合物
AU2021349316A1 (en) 2020-09-28 2023-04-27 Ascendis Pharma Bone Diseases A/S Improvement of physical and mental well-being of patients with hypoparathyroidism
AU2021405029A1 (en) 2020-12-22 2023-07-20 Technische Universität München Antibodies specific for structurally disordered sequences
CN112843222B (zh) * 2021-01-21 2023-01-31 暨南大学 Ankrd22蛋白在制备治疗或延缓自身免疫性疾病的产品中的应用
KR20230164709A (ko) 2021-04-01 2023-12-04 아센디스 파마 에이에스 염증 유발 질환을 치료하기 위한 지속형 성장 호르몬의 용도
AU2022350937A1 (en) 2021-09-22 2024-03-21 Ascendis Pharma Bone Diseases A/S Long-acting pth compound treatments
AU2022413318A1 (en) 2021-12-13 2024-05-16 Ascendis Pharma Growth Disorders A/S Effective doses of cnp conjugates
WO2023227505A1 (en) 2022-05-23 2023-11-30 Ascendis Pharma Growth Disorders A/S Liquid pharmaceutical formulations of cnp compounds
WO2024094673A1 (en) 2022-11-02 2024-05-10 Ascendis Pharma Bone Diseases A/S Pth treatment regimen comprising two pth compounds

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI92601C (fi) 1992-03-11 1994-12-12 Marja Makarow Menetelmä hyötyproteiinien erittämiseksi hiivoista
US6639050B1 (en) * 1997-07-21 2003-10-28 Ohio University Synthetic genes for plant gums and other hydroxyproline-rich glycoproteins
US20030190740A1 (en) 1998-10-13 2003-10-09 The University Of Georgia Research Foundation, Inc Stabilized bioactive peptides and methods of identification, synthesis, and use
WO2002002597A2 (en) 2000-06-30 2002-01-10 Maxygen Aps Peptide extended glycosylated polypeptides
US20020169125A1 (en) 2001-03-21 2002-11-14 Cell Therapeutics, Inc. Recombinant production of polyanionic polymers and uses thereof
US20060252120A1 (en) * 2003-05-09 2006-11-09 Kieliszewski Marcia J Synthetic genes for plant gums and other hydroxyproline-rich glycoproteins
WO2005089796A1 (en) 2004-03-15 2005-09-29 Trimeris, Inc. Site-specific chemical modification of hiv gp41-derived peptides
JP4650798B2 (ja) 2004-04-19 2011-03-16 デンカ生研株式会社 ウイルスの生産方法
CN103172747A (zh) 2005-01-25 2013-06-26 细胞治疗学公司 体内半衰期改变的生物活性蛋白质偶联物
SI2402754T2 (sl) 2006-03-06 2023-09-29 Amunix Operating Inc. Nestrukturirani rekombinantni polimeri in njihove uporabe
ES2602610T3 (es) 2007-05-31 2017-02-21 Medigene Ag Proteína estructural mutada de un parvovirus
DE602008005596D1 (de) 2007-06-21 2011-04-28 Univ Muenchen Tech Biologisch aktive proteine mit erhöhter in-vivo- und/oder in-vitro-stabilität
TW200936156A (en) * 2008-01-28 2009-09-01 Novartis Ag Methods and compositions using Klotho-FGF fusion polypeptides
MX362028B (es) 2009-02-03 2019-01-04 Amunix Pharmaceuticals Inc Polipeptidos recombinantes extendidos y composiciones que comprenden los mismos.
SG10201704777RA (en) 2009-06-08 2017-07-28 Amunix Operating Inc Growth hormone polypeptides and methods of making and using same
MX357674B (es) 2010-05-21 2018-07-18 Xl Protein Gmbh Polipeptidos biosinteticos de espira aleatoria de prolina/alanina y sus usos.

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
CA2794614A1 (en) 2011-11-24
ES2691642T3 (es) 2018-11-28
US10844094B2 (en) 2020-11-24
JP5828889B2 (ja) 2015-12-09
JP2013531480A (ja) 2013-08-08
EA024755B1 (ru) 2016-10-31
AU2011254564A1 (en) 2012-12-13
MX357674B (es) 2018-07-18
AU2011254564B2 (en) 2014-03-27
BR112012029577B1 (pt) 2022-04-12
SG185440A1 (en) 2012-12-28
US20130072420A1 (en) 2013-03-21
MX2012013286A (es) 2013-03-05
JP6038248B2 (ja) 2016-12-07
CN105477641B (zh) 2021-03-23
LT2571510T (lt) 2018-11-12
WO2011144756A1 (en) 2011-11-24
DK2571510T3 (en) 2018-11-19
BR112012029577A2 (pt) 2016-09-20
PL2571510T3 (pl) 2019-03-29
EP3473261A1 (en) 2019-04-24
CN105477641A (zh) 2016-04-13
HK1219902A1 (zh) 2017-04-21
NZ602522A (en) 2014-06-27
KR20130105289A (ko) 2013-09-25
US20160137698A1 (en) 2016-05-19
CN102883734B (zh) 2018-01-02
MX338914B (es) 2016-05-06
EA201291010A1 (ru) 2013-09-30
CA2794614C (en) 2020-12-15
US20180354992A1 (en) 2018-12-13
US10081657B2 (en) 2018-09-25
SI2571510T1 (sl) 2019-02-28
CN102883734A (zh) 2013-01-16
KR101872541B1 (ko) 2018-06-28
EP2571510A1 (en) 2013-03-27
US9221882B2 (en) 2015-12-29
JP2016026149A (ja) 2016-02-12

Similar Documents

Publication Publication Date Title
EP2571510B1 (en) Biosynthetic proline/alanine random coil polypeptides and their uses
EP2369005B1 (en) Biological active proteins having increased in vivo and/or in vitro stability
JP2022000043A (ja) Il−2部分とポリマーとのコンジュゲート
EP2855509B1 (en) Non-natural consensus albumin binding domains
EP3127923B1 (en) Method for improving solubility of protein and peptide by using immunoglobulin fc fragment linkage
JP2008543304A (ja) ヒト顆粒球コロニー刺激因子イソ型(HumanGranulocyte−ColonyStimulatingFactorIsoforms)
IL303152A (en) Compositions and methods for selective depletion of a target molecule
Choe et al. B3 (Fab)-PE38M: a recombinant immunotoxin in which a mutant form of Pseudomonas exotoxin is fused to the Fab fragment of monoclonal antibody B3
US20230312682A1 (en) SIRPalpha-FC FUSION PROTEIN
AU2021215270A1 (en) Combination therapies for delivery across the blood brain barrier
JP2023514420A (ja) 変異型rasタンパク質を標的化する分子
CN113195514A (zh) 包含混合电荷肽的融合产物和生物缀合物
Abbasi et al. Site Directed Disulfide PEGylation of Interferon-β-1b with Fork Peptide Linker
AU2013203326B2 (en) Biological active proteins having increased in vivo and/or in vitro stability
IL303149A (en) PD-L1 binding peptides and peptide complexes and methods of using them

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20121220

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20150709

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602011050803

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0038000000

Ipc: C07K0014000000

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/00 20060101AFI20170421BHEP

Ipc: C07K 7/00 20060101ALI20170421BHEP

Ipc: A61K 47/42 20170101ALI20170421BHEP

Ipc: A61K 39/00 20060101ALI20170421BHEP

Ipc: A61K 38/00 20060101ALI20170421BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20170601

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

GRAL Information related to payment of fee for publishing/printing deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

INTC Intention to grant announced (deleted)
TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

GRAL Information related to payment of fee for publishing/printing deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

INTG Intention to grant announced

Effective date: 20171215

INTC Intention to grant announced (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

INTG Intention to grant announced

Effective date: 20180622

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1026861

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180815

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011050803

Country of ref document: DE

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 2571510

Country of ref document: PT

Date of ref document: 20181029

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20180927

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: VOSSIUS AND PARTNER PATENTANWAELTE RECHTSANWAE, CH

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20181112

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2691642

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20181128

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20180808

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E016273

Country of ref document: EE

Effective date: 20181019

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181109

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181108

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011050803

Country of ref document: DE

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E041350

Country of ref document: HU

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20190509

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180808

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 1026861

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180808

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230424

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20230510

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20230505

Year of fee payment: 13

Ref country code: NO

Payment date: 20230504

Year of fee payment: 13

Ref country code: NL

Payment date: 20230510

Year of fee payment: 13

Ref country code: LT

Payment date: 20230505

Year of fee payment: 13

Ref country code: IT

Payment date: 20230505

Year of fee payment: 13

Ref country code: IE

Payment date: 20230504

Year of fee payment: 13

Ref country code: FR

Payment date: 20230508

Year of fee payment: 13

Ref country code: ES

Payment date: 20230609

Year of fee payment: 13

Ref country code: EE

Payment date: 20230523

Year of fee payment: 13

Ref country code: DK

Payment date: 20230509

Year of fee payment: 13

Ref country code: DE

Payment date: 20230531

Year of fee payment: 13

Ref country code: CZ

Payment date: 20230517

Year of fee payment: 13

Ref country code: CH

Payment date: 20230602

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230508

Year of fee payment: 13

Ref country code: SI

Payment date: 20230505

Year of fee payment: 13

Ref country code: SE

Payment date: 20230512

Year of fee payment: 13

Ref country code: PL

Payment date: 20230509

Year of fee payment: 13

Ref country code: IS

Payment date: 20230524

Year of fee payment: 13

Ref country code: HU

Payment date: 20230510

Year of fee payment: 13

Ref country code: FI

Payment date: 20230504

Year of fee payment: 13

Ref country code: AT

Payment date: 20230504

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20230510

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MT

Payment date: 20230525

Year of fee payment: 13

Ref country code: GB

Payment date: 20230525

Year of fee payment: 13